EP3912625A1 - Nouveaux ouvreurs de canaux potassiques maxi-k pour le traitement de troubles liés à l'x fragile - Google Patents

Nouveaux ouvreurs de canaux potassiques maxi-k pour le traitement de troubles liés à l'x fragile Download PDF

Info

Publication number
EP3912625A1
EP3912625A1 EP20175820.8A EP20175820A EP3912625A1 EP 3912625 A1 EP3912625 A1 EP 3912625A1 EP 20175820 A EP20175820 A EP 20175820A EP 3912625 A1 EP3912625 A1 EP 3912625A1
Authority
EP
European Patent Office
Prior art keywords
alkylene
alkyl
chloro
trifluoromethyl
methyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP20175820.8A
Other languages
German (de)
English (en)
Inventor
Roman FLECK
John Proudfoot
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Kaerus Bioscience Ltd
Original Assignee
Kaerus Bioscience Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Kaerus Bioscience Ltd filed Critical Kaerus Bioscience Ltd
Priority to EP20175820.8A priority Critical patent/EP3912625A1/fr
Priority to CA3177423A priority patent/CA3177423A1/fr
Priority to BR112022023318A priority patent/BR112022023318A2/pt
Priority to IL297493A priority patent/IL297493A/en
Priority to EP21727147.7A priority patent/EP4153170A1/fr
Priority to AU2021274879A priority patent/AU2021274879A1/en
Priority to JP2022571242A priority patent/JP2023528292A/ja
Priority to MX2022014461A priority patent/MX2022014461A/es
Priority to KR1020227044772A priority patent/KR20230016194A/ko
Priority to PCT/EP2021/063463 priority patent/WO2021234084A1/fr
Priority to CN202180035813.2A priority patent/CN115666554A/zh
Publication of EP3912625A1 publication Critical patent/EP3912625A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/424Oxazoles condensed with heterocyclic ring systems, e.g. clavulanic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/30Indoles; Hydrogenated indoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to carbon atoms of the hetero ring
    • C07D209/32Oxygen atoms
    • C07D209/34Oxygen atoms in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/30Indoles; Hydrogenated indoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to carbon atoms of the hetero ring
    • C07D209/42Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems

Definitions

  • the present invention relates to the compounds of formula (I) as defined herein below, and pharmaceutical compositions containing these compounds.
  • the compounds provided herein can act as maxi-K potassium channel openers, which makes them suitable for use in therapy, particularly in the treatment or prevention of fragile X associated disorders, such as fragile X syndrome.
  • FXS Fragile X syndrome
  • CGG trinucleotide
  • FMRP fragile X mental retardation protein
  • Maxi-K potassium channels are large-conductance, calcium (Ca 2+ )-activated potassium channels, which can conduct large amounts of potassium ions (K + ) across the cell membrane and are therefore also known as big potassium (“BK”) channels or calcium-activated big potassium (“BKCa”) channels ( Latorre R et al., Biol Res, 2006, 39(3): 385-401 ).
  • Maxi-K potassium channels (BKCa channels) are widely distributed in the brain. They function as neuronal calcium sensors and contribute to repolarization of neurons and control of neuronal signaling via regulation of cellular excitability and neurotransmitter release. In the brain, the pharmacological opening of BKCa channels can protect neurons by enhancing an endogenous neuroprotective mechanism ( Gribkoff VK et al., Nat Med, 2001, 7: 471-7 ).
  • the gene encoding BKCa ( KCNMA1 ) was identified within a de novo balanced 9q23/10q22 translocation of a patient exhibiting both autism and intellectual deficiency ( Laumonnier F et al., Am J Psychiatry, 2006, 163: 1622-9 ). Demonstration of KCNMA1 haploinsufficiency and reduced BKCa channel function in this patient suggested an underlying BKCa channelopathy behind the clinical phenotypes of autism and intellectual disability.
  • BKCa is a known molecular target of FMRP ( Deng PY et al., Neuron, 2013, 77: 696-711 ). Functionally, presynaptic BKCa channels have been shown to play an important role in FMRP regulation of action potential duration, neurotransmitter release, and short-term plasticity in hippocampal pyramidal neurons.
  • the Fmr1 knockout (KO) mouse model recapitulates many of the clinical abnormalities observed in FXS patients and is considered an appropriate model for preclinical assessment of potential therapies against FXS.
  • IP intraperitoneal
  • BMS-204352 corresponding to a C max of about 130 ng/mL
  • reversed dendritic spine abnormalities rescued hippocampal glutamate homeostasis, and reversed a range of behavioral phenotypes ( Hébert B et al., Orphanet J Rare Dis, 2014, 9: 124 ).
  • the compound BMS-204352 (MaxiPostTM), i.e. (3S)-(+)-(5-chloro-2-methoxyphenyl)-1,3-dihydro-3-fluoro-6-(trifluoromethyl)-2H-indol-2-one, is a highly potent maxi-K potassium channel opener, which was originally developed by Bristol-Myers Squibb for the treatment of acute ischemic stroke ( Jensen BS, CNS Drug Rev, 2002, 8(4): 353-60 ).
  • the present invention addresses this need and solves the problem of providing novel and improved maxi-K potassium channel openers, which have surprisingly been found to exhibit an advantageously potent stimulating activity on maxi-K channels while, at the same time, they do not give rise to metabolic degradation products that form covalent adducts with proteins (as in the case of BMS-204352) and, thus, provide an improved side effect profile and allow chronic administration. Moreover, they exhibit a beneficial solubility and pharmacokinetic profile. These properties render the compounds of the present invention particularly advantageous for use in therapy, specifically as a chronic medication for the treatment or prevention of fragile X syndrome and related disorders.
  • the present invention thus relates to a compound of the following formula (I) or a pharmaceutically acceptable salt or solvate thereof, for use in the treatment or prevention of a fragile X associated disorder, particularly fragile X syndrome.
  • R 1 is selected from C 1-5 alkyl, C 2-5 alkenyl, C 2-5 alkynyl, -(C 0-4 alkylene)-CN, -(C 1-4 alkylene)-COOH, -(C 0-4 alkylene)-COO-(C 1-5 alkyl), -(C 1-4 alkylene)-O-CO-(C 1-5 alkyl), -(C 0-4 alkylene)-CO-(C 1-5 alkyl), -(C 0-4 alkylene)-CO-NH 2 , -(C 0-4 alkylene)-CO-NH(C 1-5 alkyl), -(C 0-4 alkylene)-CO-N(C 1-5 alkyl)(C 1-5 alkyl), -(C 0-4 alkylene)-NH-CO-(C 1-5 alkyl), -(C 0-4 alkylene)-N(C 1-5 alkyl)-CO-(C 1-5 alkyl),
  • R 2 is hydrogen
  • Ring A is phenyl or a 5- or 6-membered monocyclic heteroaryl, wherein said phenyl or said heteroaryl is fused to the ring containing -CO-NR 2 -.
  • Each R 3 is independently selected from C 1-5 alkyl, C 2-5 alkenyl, C 2-5 alkynyl, -(C 0-4 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-O(C 1-5 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-SH, -(C 0-4 alkylene)-S(C 1-5 alkyl), -(C 0-4 alkylene)-NH 2 , -(C 0-4 alkylene)-NH(C 1-5 alkyl), -(C 0-4 alkylene)-N(C 1-5 alkyl)(C 1-5 alkyl), -(C 0-4 alkylene)-NH-OH, -(C 0-4 alkylene)-N(C 1
  • n is an integer of 0 to 4.
  • the ring atoms Z 1 , Z 2 and Z 3 are each independently C(-R 6 ) or N.
  • R 4 is selected from hydrogen, C 1-5 alkyl, C 2-5 alkenyl, C 2-5 alkynyl, -(C 0-4 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-O(C 1-5 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-SH, -(C 0-4 alkylene)-S(C 1-5 alkyl), -(C 0-4 alkylene)-NH 2 , -(C 0-4 alkylene)-NH(C 1-5 alkyl), -(C 0-4 alkylene)-N(C 1-5 alkyl)(C 1-5 alkyl), -(C 0-4 alkylene)-NH-OH, -(C 0-4 alkylene)-N(C 1
  • R 5A is selected from hydrogen, C 1-5 alkyl, C 2-5 alkenyl, C 2-5 alkynyl, -(C 0-4 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-O(C 1-5 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-SH, -(C 0-4 alkylene)-S(C 1-5 alkyl), -(C 0-4 alkylene)-NH 2 , -(C 0-4 alkylene)-NH(C 1-5 alkyl), -(C 0-4 alkylene)-N(C 1-5 alkyl)(C 1-5 alkyl), -(C 0-4 alkylene)-NH-OH, -(C 0-4 alkylene)-N(C
  • Each R 6 is independently selected from hydrogen, C 1-5 alkyl, C 2-5 alkenyl, C 2-5 alkynyl, -(C 0-4 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-O(C 1-5 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-SH, -(C 0-4 alkylene)-S(C 1-5 alkyl), -(C 0-4 alkylene)-NH 2 , -(C 0-4 alkylene)-NH(C 1-5 alkyl), -(C 0-4 alkylene)-N(C 1-5 alkyl)(C 1-5 alkyl), -(C 0-4 alkylene)-NH-OH, -(C 0-4 alkylene)-N(
  • Each R Cyc is independently selected from C 1-5 alkyl, C 2-5 alkenyl, C 2-5 alkynyl, -(C 0-4 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-O(C 1-5 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-SH, -(C 0-4 alkylene)-S(C 1-5 alkyl), -(C 0-4 alkylene)-NH 2 , -(C 0-4 alkylene)-NH(C 1-5 alkyl), -(C 0-4 alkylene)-N(C 1-5 alkyl)(C 1-5 alkyl), -(C 0-4 alkylene)-NH-OH, -(C 0-4 alkylene)-N(C
  • the present invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient, for use in the treatment or prevention of a fragile X associated disorder (particularly fragile X syndrome).
  • the invention in particular provides a pharmaceutical composition comprising, as an active ingredient, a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof, together with a pharmaceutically acceptable excipient, for use in the treatment or prevention of a fragile X associated disorder (such as fragile X syndrome).
  • the invention also relates to the use of a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof in the preparation of a medicament (or pharmaceutical composition) for the treatment or prevention of a fragile X associated disorder.
  • the present invention likewise relates to a method of treating or preventing a fragile X associated disorder (particularly fragile X syndrome), the method comprising administering a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising any of the aforementioned entities in combination with a pharmaceutically acceptable excipient, to a subject (preferably a human) in need thereof.
  • a therapeutically effective amount of the compound of formula (I) or the pharmaceutically acceptable salt or solvate thereof (or of the pharmaceutical composition) is to be administered in accordance with this method.
  • the fragile X associated disorder to be treated or prevented in accordance with the present invention may be, in particular, a genetic disease/disorder caused by (or associated with) a change or mutation in the fragile X mental retardation 1 ( FMR1 ) gene and/or in the fragile X mental retardation 2 ( FMR2 ) gene, such as a pathologically enhanced number of CGG repeats (e.g., the presence of about 45 or more CGG repeats, particularly about 55 or more CGG repeats, especially more than about 200 CGG repeats, or even more than about 230 CGG repeats in the FMR1 gene), or a disease/disorder caused by (or associated with) a deficiency, absence, functional impairment, or dysfunction of the protein encoded by the FMR1 gene, i.e.
  • a genetic disease/disorder caused by (or associated with) a change or mutation in the fragile X mental retardation 1 ( FMR1 ) gene and/or in the fragile X mental retardation 2 ( FMR2 ) gene such
  • fragment X associated disorder is used herein synonymously and interchangeably with “fragile X spectrum disorder”.
  • the fragile X associated disorder to be treated or prevented in accordance with the present invention is selected from fragile X syndrome (FXS), fragile X-associated tremor/ataxia syndrome (FXTAS), fragile X-associated primary ovarian insufficiency (FXPOI), fragile X-associated polycystic ovarian syndrome, fragile XE-associated mental retardation (FRAXE), a clinical syndrome associated with pathogenic mutations in the FMR1 gene (particularly such mutations that result in altered BK channel function; see, e.g., Myrick LK et al., Proc Natl Acad Sci USA, 2015, 112(4): 949-56 ), and a clinical syndrome associated with pathogenic variation in the KCNMA1 gene (such as, e.g., paroxysmal nonkinesigenic dyskinesia (PNKD), particularly PNKD3, with or without generalized epilepsy; see, e.g., Liang L et al., Hum Mol Genet,
  • the fragile X associated disorder is selected from fragile X syndrome (FXS), fragile X-associated tremor/ataxia syndrome (FXTAS), fragile X-associated primary ovarian insufficiency (FXPOI), fragile X-associated polycystic ovarian syndrome, and fragile XE-associated mental retardation (FRAXE).
  • FXS fragile X syndrome
  • FXTAS fragile X-associated tremor/ataxia syndrome
  • FXPOI fragile X-associated primary ovarian insufficiency
  • FXE-associated mental retardation FAXE
  • the fragile X associated disorder is selected from fragile X syndrome (FXS), fragile X-associated tremor/ataxia syndrome (FXTAS), and fragile X-associated primary ovarian insufficiency (FXPOI).
  • the fragile X associated disorder is fragile X syndrome.
  • the present invention relates, in particular, to a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof (or a pharmaceutical composition comprising any of the aforementioned entities in combination with a pharmaceutically acceptable excipient) for use in the treatment or prevention of fragile X syndrome (FXS), fragile X-associated tremor/ataxia syndrome (FXTAS), or fragile X-associated primary ovarian insufficiency (FXPOI), preferably for use in the treatment or prevention of fragile X syndrome.
  • FXS fragile X syndrome
  • FXTAS fragile X-associated tremor/ataxia syndrome
  • FXPOI fragile X-associated primary ovarian insufficiency
  • the present invention furthermore relates to the use of a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof (as defined in the first aspect of the invention) as a maxi-K potassium channel opener in research, particularly as a research tool compound for stimulating maxi-K potassium channels.
  • the invention refers to the in vitro use of a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof as a maxi-K potassium channel opener and, in particular, to the in vitro use of a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof as a research tool compound acting as a maxi-K potassium channel opener.
  • the invention likewise relates to a method, particularly an in vitro method, of stimulating maxi-K potassium channels, the method comprising the application of a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof.
  • the invention further relates to a method of stimulating maxi-K potassium channels, the method comprising applying a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof to a test sample (e.g., a biological sample) or a test animal (i.e., a non-human test animal).
  • the invention also refers to a method, particularly an in vitro method, of stimulating maxi-K potassium channels in a sample (e.g., a biological sample), the method comprising applying a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof to said sample.
  • the present invention further provides a method of stimulating maxi-K potassium channels, the method comprising contacting a test sample (e.g., a biological sample) or a test animal (i.e., a non-human test animal) with a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof.
  • sample includes, without being limited thereto: a cell, a cell culture, or a cellular or subcellular extract; biopsied material obtained from an animal (e.g., a human), or an extract thereof; or blood, serum, plasma, saliva, urine, or any other body fluid, or an extract thereof.
  • in vitro is used in this specific context in the sense of "outside a living human or animal body", which includes, in particular, experiments performed with cells, cellular or subcellular extracts, and/or biological molecules in an artificial environment such as an aqueous solution or a culture medium which may be provided, e.g., in a flask, a test tube, a Petri dish, a microtiter plate, etc.
  • the present invention provides novel compounds, i.e., the invention relates to a compound of the following formula (Ia) or a pharmaceutically acceptable salt or solvate thereof: wherein the groups/variables in formula (Ia), including in particular ring A, Z 1 , Z 2 , Z 3 , R 1 , R 2 , R 3 , R 4 , R 5 , R 5A , R 6 , R Cyc and n, have the same meanings, including the same preferred meanings, as the corresponding groups/variables comprised in the compound of formula (I) according to the first aspect of the invention; and wherein the following further conditions apply to the compound of formula (Ia):
  • the compounds of formula (Ia) can be used as active pharmaceutical ingredients, particularly as maxi-K potassium channel openers.
  • the present invention thus also relates to the compound of formula (Ia) or a pharmaceutically acceptable salt or solvate thereof for use in the treatment or prevention of a fragile X associated disorder (such as, e.g., fragile X syndrome (FXS), fragile X-associated tremor/ataxia syndrome (FXTAS), or fragile X-associated primary ovarian insufficiency (FXPOI)) or other diseases/disorders in which maxi-K potassium channels are implicated.
  • a fragile X associated disorder such as, e.g., fragile X syndrome (FXS), fragile X-associated tremor/ataxia syndrome (FXTAS), or fragile X-associated primary ovarian insufficiency (FXPOI)
  • FXPOI fragile X-associated primary ovarian insufficiency
  • the present invention further relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (Ia), or a pharmaceutically acceptable salt or solvate thereof, in combination with a pharmaceutically acceptable excipient.
  • the invention relates to a compound of formula (Ia) or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising any of the aforementioned entities and a pharmaceutically acceptable excipient, for use as a medicament.
  • the invention refers to a compound of formula (Ia) or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising any of the aforementioned entities and a pharmaceutically acceptable excipient, for use in the treatment or prevention of a disease/disorder selected from stroke (e.g., ischemic stroke), ischemia reperfusion injury, ischemic heart disease, hypertension (e.g., partial hypertension or arterial hypertension), myocardial infarction, allergic rhinitis, asthma, chronic obstructive pulmonary disease (COPD), multiple sclerosis, spasticity (e.g., muscle spasticity, or spasticity in a subject/patient suffering from multiple sclerosis), tremor, a muscular disorder, dyskinesia, bladder dysfunction, overactive bladder (OAB), urinary incontinence, irritable bowel syndrome (IBS), faecal incontinence, constipation, gastro-oesophageal reflux disorder (GERD), impaired gastrointenst
  • the invention also relates to the use of a compound of formula (Ia) or a pharmaceutically acceptable salt or solvate thereof in the preparation of a medicament (or pharmaceutical composition) for the treatment or prevention of a disease/disorder selected from stroke (e.g., ischemic stroke), ischemia reperfusion injury, ischemic heart disease, hypertension (e.g., partial hypertension or arterial hypertension), myocardial infarction, allergic rhinitis, asthma, chronic obstructive pulmonary disease (COPD), multiple sclerosis, spasticity (e.g., muscle spasticity, or spasticity in a subject/patient suffering from multiple sclerosis), tremor, a muscular disorder, dyskinesia, bladder dysfunction, overactive bladder (OAB), urinary incontinence, irritable bowel syndrome (IBS), faecal incontinence, constipation, gastro-oesophageal reflux disorder (GERD), impaired gastrointenstinal passage, detrus
  • stroke e.g
  • the invention likewise relates to a method of treating or preventing a disease/disorder selected from stroke (e.g., ischemic stroke), ischemia reperfusion injury, ischemic heart disease, hypertension (e.g., partial hypertension or arterial hypertension), myocardial infarction, allergic rhinitis, asthma, chronic obstructive pulmonary disease (COPD), multiple sclerosis, spasticity (e.g., muscle spasticity, or spasticity in a subject/patient suffering from multiple sclerosis), tremor, a muscular disorder, dyskinesia, bladder dysfunction, overactive bladder (OAB), urinary incontinence, irritable bowel syndrome (IBS), faecal incontinence, constipation, gastro-oesophageal reflux disorder (GERD), impaired gastrointenstinal passage, detrusor underactivity, erectile dysfunction, opioid-induced respiratory depression, anxiety, an anxiety disorder (e.g., generalized anxiety disorder (GAD) or
  • R 1 is selected from C 1-5 alkyl, C 2-5 alkenyl, C 2-5 alkynyl, -(C 0-4 alkylene)-CN (e.g., -CH 2 -CN), -(C 1-4 alkylene)-COOH (e.g., -CH 2 -COOH), -(C 0-4 alkylene)-COO-(C 1-5 alkyl), -(C 1-4 alkylene)-O-CO-(C 1-5 alkyl), -(C 0-4 alkylene)-CO-(C 1-5 alkyl), -(C 0-4 alkylene)-CO-NH 2 , -(C 0-4 alkylene)-CO-NH(C 1-5 alkyl), -(C 0-4 alkylene)-CO-N(C 1-5 alkyl)(C 1-5 alkyl), -(C 0-4 alkylene)-NH-CO-(C 1-5 alkyl), -
  • R 1 is C 1-5 alkyl, and R 5 is R 5A ; or alternatively, R 1 and R 5 are mutually joined to form a group -CH 2 CH 2 -, -O-CH 2 -, -CH 2 -O-, -CO-O- or -O-CO-. More preferably, R 1 is C 1-5 alkyl, and R 5 is R 5A . Even more preferably, R 1 is methyl or ethyl, and R 5 is R 5A . Yet even more preferably, R 1 is methyl, and R 5 is R 5A .
  • R 1 and R 5 may also be mutually joined to form a C 2-3 alkylene or a C 2-3 alkenylene, wherein one or two -CH 2 - units in said C 2-3 alkylene or said C 2-3 alkenylene are each optionally replaced by a group independently selected from -O-, -NH-, -N(C 1-5 alkyl)-, and -CO-.
  • the C 2-3 alkylene or the C 2-3 alkenylene may be a linear C 2-3 alkylene or a linear C 2-3 alkenylene group, which is attached via its distal ends (i.e., via the two carbon atoms at the ends/termini of the alkylene or alkenylene chain) to the ring atoms carrying R 1 and R 5 , respectively.
  • R 1 and R 5 are mutually joined to form a group -CH 2 CH 2 - or -CH 2 CH 2 CH 2 -, wherein one or two -CH 2 - units in said -CH 2 CH 2 - or -CH 2 CH 2 CH 2 -are each optionally replaced by a group independently selected from -O-, -NH-, -N(C 1-5 alkyl)-, and -CO-.
  • R 1 and R 5 may be mutually joined to form a group -CH 2 CH 2 -, -O-CH 2 -, -CH 2 -O-, -CO-O-, or -O-CO-.
  • R 1 is C 1-5 alkyl (particularly methyl), and R 5 is R 5A .
  • R 2 is hydrogen
  • ring A is selected from phenyl, pyridinyl, oxazolyl, and isoxazolyl, wherein said phenyl, said pyridinyl, said oxazolyl or said isoxazolyl is fused to the ring containing -CO-NR 2 -. More preferably, ring A is phenyl or pyridinyl, wherein said phenyl or said pyridinyl is fused to the ring containing -CO-NR 2 -. If ring A is pyridinyl (which is fused to the ring containing -CO-NR 2 -), then the compound of formula (I) may have one of the following structures:
  • ring A is phenyl which is fused to the ring containing -CO-NR 2 -. Accordingly, it is particularly preferred that the compound of formula (I) has the following structure:
  • Each R 3 is independently selected from C 1-5 alkyl, C 2-5 alkenyl, C 2-5 alkynyl, -(C 0-4 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-O(C 1-5 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-SH, -(C 0-4 alkylene)-S(C 1-5 alkyl), -(C 0-4 alkylene)-NH 2 , -(C 0-4 alkylene)-NH(C 1-5 alkyl), -(C 0-4 alkylene)-N(C 1-5 alkyl)(C 1-5 alkyl), -(C 0-4 alkylene)-NH-OH, -(C 0-4 alkylene)-N(C 1
  • each R 3 is independently selected from C 1-5 alkyl, C 2-5 alkenyl, C 2-5 alkynyl, -OH, -O(C 1-5 alkyl), -O(C 1-5 alkylene)-OH, -O(C 1-5 alkylene)-O(C 1-5 alkyl), -SH, -S(C 1-5 alkyl), -NH 2 , -NH(C 1-5 alkyl), -N(C 1-5 alkyl)(C 1-5 alkyl), -NH-OH, -N(C 1-5 alkyl)-OH, -NH-O(C 1-5 alkyl), -N(C 1-5 alkyl)-O(C 1-5 alkyl), halogen, C 1-5 haloalkyl, -O-(C 1-5 haloalkyl), -CN, -CHO, -CO-(C 1-5 alkyl), -COOH, -CO-O-(C 1-5 alkyl),
  • each R 3 is independently selected from C 1-5 alkyl, -OH, -O(C 1-5 alkyl), -O(C 1-5 alkylene)-OH, -O(C 1-5 alkylene)-O(C 1-5 alkyl), -SH, -S(C 1-5 alkyl), -NH 2 , -NH(C 1-5 alkyl), -N(C 1-5 alkyl)(C 1-5 alkyl), halogen, C 1-5 haloalkyl, -O-(C 1-5 haloalkyl), -CN, and C 3-7 cycloalkyl.
  • each R 3 is independently selected from C 1-5 alkyl, -O(C 1-5 alkyl), halogen, C 1-5 haloalkyl, and -CN. Yet even more preferably, each R 3 is independently halogen (e.g., -F, -Cl or -Br) or C 1-5 haloalkyl (e.g., -CF 3 , -CHF 2 or -CH 2 F). Still more preferably, each R 3 is -CF 3 .
  • n is an integer of 0 to 4 (i.e., 0, 1, 2, 3 or 4). Preferably, n is 0, 1 or 2. More preferably, n is 1.
  • n indicates the number of substituents R 3 that are bound to ring A. If n is 0, then ring A is not substituted with any group R 3 , i.e. is substituted with hydrogen instead of R 3 .
  • the group R 3 (if n is 1), or at least one group R 3 (if n is 2, 3 or 4), is C 1-5 haloalkyl (e.g., -CF 3 , -CHF 2 or -CH 2 F) or halogen (e.g., -F, -Cl or -Br), even more preferably -CF 3 .
  • haloalkyl e.g., -CF 3 , -CHF 2 or -CH 2 F
  • halogen e.g., -F, -Cl or -Br
  • said C 1-5 haloalkyl or said halogen is attached to ring A (if ring A is a six-membered ring, such as phenyl or pyridinyl) at a ring carbon atom in the 4-position, the 5-position, the 6-position or the 7-position, more preferably in the 5-position, the 6-position or the 7-position, even more preferably in the 5-position or the 6-position, yet even more preferably in the 6-position, wherein the numbering of ring atoms is as illustrated in the following:
  • the ring atoms Z 1 , Z 2 and Z 3 are each independently C(-R 6 ) or N.
  • two of the ring atoms Z 1 , Z 2 and Z 3 are each independently C(-R 6 ) or N, and the other one is C(-R 6 ).
  • Z 1 and Z 2 may each be N
  • Z 3 may be C(-R 6 ).
  • one of the ring atoms Z 1 , Z 2 and Z 3 is C(-R 6 ) or N
  • the other two ring atoms are each C(-R 6 ).
  • one of the ring atoms Z 1 and Z 2 is C(-R 6 ) or N
  • the other one of said ring atoms Z 1 and Z 2 is C(-R 6 )
  • Z 3 is C(-R 6 ).
  • Z 2 is C(-R 6 ) or N, while Z 1 and Z 3 are each C(-R 6 ).
  • Z 2 may be N, while Z 1 and Z 3 may each be C(-R 6 ); in this case, the compound of formula (I) has the following structure:
  • Z 1 , Z 2 and Z 3 are each C(-R 6 ). Accordingly, it is particularly preferred that the compound of formula (I) has the following structure:
  • R 4 is selected from hydrogen, C 1-5 alkyl, C 2-5 alkenyl, C 2-5 alkynyl, -(C 0-4 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-O(C 1-5 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-SH, -(C 0-4 alkylene)-S(C 1-5 alkyl), -(C 0-4 alkylene)-NH 2 , -(C 0-4 alkylene)-NH(C 1-5 alkyl), -(C 0-4 alkylene)-N(C 1-5 alkyl)(C 1-5 alkyl), -(C 0-4 alkylene)-NH-OH, -(C 0-4 alkylene)-N(C 1
  • R 4 is selected from hydrogen, C 1-5 alkyl, -O(C 1-5 alkyl), halogen, C 1-5 haloalkyl, -O(C 1-5 haloalkyl) (e.g., -OCF 3 ), -CN, -SO 2 -(C 1-5 alkyl), -(C 0-4 alkylene)-carbocyclyl, and -(C 0-4 alkylene)-heterocyclyl (such as -(C 0-4 alkylene)-heteroaryl; e.g., imidazolyl), wherein the carbocyclyl moiety in said -(C 0-4 alkylene)-carbocyclyl and the heterocyclyl moiety in said -(C 0-4 alkylene)-heterocyclyl are each optionally substituted with one or more groups R Cyc .
  • R 4 is selected from hydrogen, C 1-5 alkyl, -O(C 1-5 alkyl), halogen, C 1-5 haloalkyl, -O(C 1-5 haloalkyl) (e.g., -OCF 3 ), -CN, -SO 2 -(C 1-5 alkyl), aryl, and heteroaryl (such as, e.g., imidazolyl), wherein said aryl and said heteroaryl are each optionally substituted with one or more groups R CYc (for example, R 4 may be N-methylimidazolyl, such as 1-methylimidazol-2-yl, 1-methylimidazol-4-yl, or 1-methylimidazol-5-yl). Even more preferably, R 4 is halogen (e.g., -F, -Cl, -Br or -I), -CF 3 , or -CN. It is particularly preferred that R 4 is -Cl.
  • R 5 is either a group R 5A ; or R 1 and R 5 are mutually joined to form a C 2-3 alkylene or a C 2-3 alkenylene, wherein one or two -CH 2 - units in said C 2-3 alkylene or said C 2-3 alkenylene are each optionally replaced by a group independently selected from -O-, -NH-, -N(C 1-5 alkyl)-, and -CO-.
  • R 5 is a group R 5A .
  • R 5A is selected from hydrogen, C 1-5 alkyl, C 2-5 alkenyl, C 2-5 alkynyl, -(C 0-4 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-O(C 1-5 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-SH, -(C 0-4 alkylene)-S(C 1-5 alkyl), -(C 0-4 alkylene)-NH 2 , -(C 0-4 alkylene)-NH(C 1-5 alkyl), -(C 0-4 alkylene)-N(C 1-5 alkyl)(C 1-5 alkyl), -(C 0-4 alkylene)-NH-OH, -(C 0-4 alkylene)-N(C
  • R 5A is selected from hydrogen, -O(C 1-5 alkyl), -OH, halogen, C 1-5 haloalkyl, -O-(C 1-5 haloalkyl), -CN, -CO-(C 1-5 alkyl), -COOH, -CO-O-(C 1-5 alkyl), -CO-NH 2 , -CO-NH(C 1-5 alkyl), and -CO-N(C 1-5 alkyl)(C 1-5 alkyl).
  • R 5A is selected from hydrogen, -O(C 1-5 alkyl), -OH, halogen, C 1-5 haloalkyl, -CN, -COOH, and -CO-NH 2 . Even more preferably, R 5A is -O(C 1-5 alkyl) or -OH. Yet even more preferably, R 5A is -OCH 3 , -OCH 2 CH 3 or -OH. Still more preferably, R 5A is -OCH 3 .
  • Each R 6 is independently selected from hydrogen, C 1-5 alkyl, C 2-5 alkenyl, C 2-5 alkynyl, -(C 0-4 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-O(C 1-5 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-SH, -(C 0-4 alkylene)-S(C 1-5 alkyl), -(C 0-4 alkylene)-NH 2 , -(C 0-4 alkylene)-NH(C 1-5 alkyl), -(C 0-4 alkylene)-N(C 1-5 alkyl)(C 1-5 alkyl), -(C 0-4 alkylene)-NH-OH, -(C 0-4 alkylene)-N(
  • each R 6 is independently selected from hydrogen, C 1-5 alkyl, C 2-5 alkenyl, C 2-5 alkynyl, -OH, -O(C 1-5 alkyl), -O(C 1-5 alkylene)-OH, -O(C 1-5 alkytene)-O(C 1-5 alkyl), -SH, -S(C 1-5 alkyl), -NH 2 , -NH(C 1-5 alkyl), -N(C 1-5 alkyl)(C 1-5 alkyl), -NH-OH, -N(C 1-5 alkyl)-OH, -NH-O(C 1-5 alkyl), -N(C 1-5 alkyl)-O(C 1-5 alkyl), halogen, C 1-5 haloalkyl, -O-(C 1-5 haloalkyl), -CN, -CHO, -CO-(C 1-5 alkyl), -COOH, -CO-O-(C
  • each R 6 is independently selected from hydrogen, C 1-5 alkyl, -O(C 1-5 alkyl), halogen, C 1-5 haloalkyl, -O-(C 1-5 haloalkyl), and -CN. Even more preferably, each R 6 is hydrogen.
  • Each R Cyc is independently selected from C 1-5 alkyl, C 2-5 alkenyl, C 2-5 alkynyl, -(C 0-4 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-O(C 1-5 alkylene)-OH, -(C 0-4 alkylene)-O(C 1-5 alkylene)-O(C 1-5 alkyl), -(C 0-4 alkylene)-SH, -(C 0-4 alkylene)-S(C 1-5 alkyl), -(C 0-4 alkylene)-NH2, -(C 0-4 alkylene)-NH(C 1-5 alkyl), -(C 0-4 alkylene)-N(C 1-5 alkyl)(C 1-5 alkyl), -(C 0-4 alkylene)-NH-OH, -(C 0-4 alkylene)-N(C 1-5
  • each R Cyc is independently selected from C 1-5 alkyl, C 2-5 alkenyl, C 2-5 alkynyl, -OH, -O(C 1-5 alkyl), -O(C 1-5 alkylene)-OH, -O(C 1-5 alkylene)-O(C 1-5 alkyl), -SH, -S(C 1-5 alkyl), -NH 2 , -NH(C 1-5 alkyl), -N(C 1-5 alkyl)(C 1-5 alkyl), -NH-OH, -N(C 1-5 alkyl)-OH, -NH-O(C 1-5 alkyl), -N(C 1-5 alkyl)-O(C 1-5 alkyl), halogen, C 1-5 haloalkyl, -O-(C 1-5 haloalkyl), -CN, -CHO, -CO-(C 1-5 alkyl), -COOH, -CO-O-(C 1-5 alkyl
  • each R Cyc is independently selected from C 1-5 alkyl, -OH, -O(C 1-5 alkyl), -O(C 1-5 alkylene)-OH, -O(C 1-5 alkylene)-O(C 1-5 alkyl), -SH, -S(C 1-5 alkyl), -NH 2 , -NH(C 1-5 alkyl), -N(C 1-5 alkyl)(C 1-5 alkyl), halogen, C 1-5 haloalkyl, -O-(C 1-5 haloalkyl), and -CN.
  • the compound of formula (I) is any one of the specific compounds of formula (I) described in the examples section of this specification, including any one of Examples 1 to 40 described further below, either in non-salt form or as a pharmaceutically acceptable salt or solvate of the respective compound.
  • the compound of formula (I) is selected from: 3-(5-chloro-2-methoxyphenyl)-3-methyl-6-(trifluoromethyl)indolin-2-one; 3-(5-chloro-2-hydroxy-phenyl)-3-methyl-6-(trifluoromethyl)indolin-2-one; 3-(5-chloro-2-methoxyphenyl)-3-ethyl-6-(trifluoromethyl)indolin-2-one; 3-(5-chloro-2-methoxyphenyl)-3-propyl-6-(trifluoromethyl)indol in-2-one; 3-benzyl-3-(5-chloro-2-methoxyphenyl)-6-(trifluoromethyl)indolin-2-one; 2-(3-(5-chloro-2-methoxyphenyl)-2-oxo-6-(trifluoromethyl)indolin-3-yl)acetonitrile; 3-allyl-3-(
  • the invention furthermore relates specifically and individually to each stereoisomer of the above-mentioned compounds, including in particular to each one of these compounds having the S-configuration at the carbon atom carrying R 1 , or to each one of these compounds having the R-configuration at the carbon atom carrying R 1 .
  • the present invention also provides novel compounds. Accordingly, in the above-discussed third aspect, the invention relates to a compound of the following formula (Ia) or a pharmaceutically acceptable salt or solvate thereof.
  • the compounds of formula (Ia) can be used as active pharmaceutical ingredients, particularly as maxi-K potassium channel openers.
  • the present invention thus also relates to the compound of formula (Ia) or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising any of the aforementioned entities and a pharmaceutically acceptable excipient, for use in the treatment or prevention of a fragile X associated disorder (such as, e.g., fragile X syndrome (FXS), fragile X-associated tremor/ataxia syndrome (FXTAS), or fragile X-associated primary ovarian insufficiency (FXPOI)) or other diseases/disorders in which maxi-K potassium channels are implicated.
  • a fragile X associated disorder such as, e.g., fragile X syndrome (FXS), fragile X-associated tremor/ataxia syndrome (FXTAS), or fragile X-associated primary ovarian insufficiency (FXPOI)
  • FXPOI fragile X-
  • the invention refers to a compound of formula (Ia) or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising any of the aforementioned entities and a pharmaceutically acceptable excipient, for use in the treatment or prevention of a disease/disorder selected from stroke (e.g., ischemic stroke), ischemia reperfusion injury, ischemic heart disease, hypertension (e.g., partial hypertension or arterial hypertension), myocardial infarction, allergic rhinitis, asthma, chronic obstructive pulmonary disease (COPD), multiple sclerosis, spasticity (e.g., muscle spasticity, or spasticity in a subject/patient suffering from multiple sclerosis), tremor, a muscular disorder, dyskinesia, bladder dysfunction, overactive bladder (OAB), urinary incontinence, irritable bowel syndrome (IBS), faecal incontinence, constipation, gastro-oesophageal reflux disorder (GERD), impaired gastrointenst
  • the groups/variables comprised in formula (Ia), including in particular ring A, Z 1 , Z 2 , Z 3 , R 1 , R 2 , R 3 , R 4 , R 5 , R 5A , R 6 , R Cyc and n, have the same meanings, including the same preferred meanings, as described and defined herein above in relation with the compound of formula (I).
  • ring A is phenyl (which is fused to the ring containing -CO-NR 2 -), n is 1, 2 or 3, and a group R 3 is attached to ring A in 5-position (wherein the numbering of ring atoms is as illustrated below), then it is preferred that said group R 3 which is attached in 5-position is not -Br; more preferably, said group R 3 which is attached in 5-position is not halogen (i.e., is different from halogen):
  • ring A, Z 1 , Z 2 , Z 3 , R 1 , R 2 , R 3 , R 4 , R 5 , R 5A , R 6 , R Cyc , and n comprised in the compound of formula (Ia) according to the third aspect of the invention have the same preferred meanings as described and defined herein above in relation to the compound of formula (I) according to the first aspect of the invention.
  • the compound of formula (Ia) is any one of the corresponding specific compounds described in the examples section of this specification, either in non-salt form or as a pharmaceutically acceptable salt or solvate of the respective compound.
  • the compound of formula (Ia) is selected from:
  • the invention also relates specifically and individually to each stereoisomer of the above-mentioned compounds, including in particular to each one of these compounds having the S-configuration at the carbon atom carrying R 1 , or to each one of these compounds having the R-configuration at the carbon atom carrying R 1 .
  • the compound of formula (Ia) is selected from:
  • the compounds of formula (I) or (Ia) and their pharmaceutically acceptable salts or solvates will be readily apparent.
  • the compounds of the invention can be prepared in accordance with, or in analogy to, the synthetic routes described herein below in the examples section.
  • the compounds of formula (I) or (Ia) can be synthesized in accordance with the general methods described in the following Schemes 1 to 8.
  • Step 1 To a mixture of phenylacetate ester (1, 5 mmol, 1 eq) and the 1-fluoro-2-nitrobenzene derivative (1.2 eq ) in an inert solvent such as THF (40 mL) cooled to -40°C is added a base such as LiHMDS (1.3 eq). The mixture is warmed to ambient temperature and stirred until the reaction is complete. The reaction is quenched by addition of saturated aqueous ammonium chloride and extracted with ethyl acetate. The organic phase is washed with saturated brine, dried over anhydrous sodium sulfate, filtered and evaporated. Chromatography of the residue over silica gel (eluent petroleum ether / ethyl acetate gradient) gives the product 2.
  • Step 2 To a stirred solution of 2 (1 mmol, 1 eq) and alkyl or aryl halide, R 1 -X (6 eq) in an inert solvent such as DMF (5 mL), cooled on ice, is added a deprotonating agent such as NaH (1.5 eq). The mixture is stirred at on ice for 0.5 hr, then warmed to 50 °C and stirred until the reaction is complete. The reaction is cooled on ice, quenched with water and extracted with EtOAc. The organic phase is washed with saturated brine, dried over anhydrous sodium sulfate, filtered and evaporated. Chromatography of the residue over silica gel (eluent petroleum ether / ethyl acetate gradient) gives the product 3.
  • a deprotonating agent such as NaH (1.5 eq).
  • Step 3 To a solution of 3 (0.5 mmol, 1 eq) in mixture of ethanol / water is added a reducing agent such as sodium dithionite (10 eq ) and the mixture is stirred at 80 °C until the reaction is complete. The reaction mixture is filtered and concentrated under reduced pressure and the residue is purified by chromatography over silica gel (eluent petroleum ether / ethyl acetate gradient) to give the product 4.
  • a reducing agent such as sodium dithionite (10 eq )
  • Step 1 To a solution of ethyl 2-(heteroaryl)acetate 1 (10 mmol, 1 eq) in THF (10 mL) is added dropwise a deprotonating agent such as NaH (1 eq) at -50 °C under N 2 . The mixture is stirred for 1 h. The alkyl iodide, R 1 -X, (5 eq) is added slowly and the reaction mixture is warmed to -10 °C, and stirred until the reaction is complete. Water (20 mL) is added at 0°C, and the mixture is extracted with EtOAc. The organic phase is washed saturated brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue is purified by chromatography to give the product 2.
  • a deprotonating agent such as NaH (1 eq) at -50 °C under N 2 .
  • Step 2 To a solution of the ethyl 2-(heteroaryl)acetate 2 (5 mmol, 1 eq) and the 1-fluoro-2-nitrobenzene derivative (1.02 eq) in THF (5 mL) is added a deprotonating agent such as KOtBu (1.5 eq). The mixture is stirred at 25 °C until the reaction is complete. The reaction is quenched by addition saturated NH 4 Cl at 0°C and extracted with EtOAc. The organic phase is washed with saturated brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue is purified by chromatography to give product 3.
  • a deprotonating agent such as KOtBu (1.5 eq).
  • Step 3 To a solution of 3 (3 mmol, 1 eq) in AcOH (10 mL) is added Fe (10 eq) and the mixture is stirred at 50 °C for until the reaction is complete. The mixture is filtered, concentrated under reduced pressure and the residue is purified by chromatography to give the product 4.
  • Step 1 To a mixture of ethyl phenylacetate derivative 1 (5 mmol, 1 eq) and 2-chloro-3-nitropyridine derivative (1.02 eq) in THF (10 mL) is added a deprotonating agent such as t-BuOK (1.5 eq) at 0°C under N 2 . The mixture is stirred at 25°C until the reaction is complete. The mixture is filtered and concentrated under reduced pressure and the residue is purified by chromatography to give product 2 .
  • a deprotonating agent such as t-BuOK (1.5 eq)
  • Step 2 To a mixture of 2 (1 mmol, 1 eq) and Mel (6 eq) in DMF (5 mL) is added a deprotonating agent such as NaH (1.5 eq) at 0°C under N 2 . The mixture is stirred at 0 °C for 0.5 h and then heated to 50 °C and stirred until the reaction is complete. The reaction is quenched with water, extracted with EtOAc, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue is purified by chromatography to give product 3.
  • a deprotonating agent such as NaH (1.5 eq)
  • Step 3 To a mixture of 3 (1 mmol, 1 eq) in AcOH (5 mL) is added Fe (10 eq) in one portion at 25°C under N 2 . The mixture is stirred at 60 °C until the reaction is complete. The mixture is filtered and concentrated under reduced pressure. The residue is purified by chromatography to give the product 4.
  • Step 1 To a solution of hydroxybenzaldehyde derivative 1 (50 mmol, 1 eq) in DCM (150 mL) is added BF 3 .Et 2 O (0.1 eq) and the mixture is stirred at 15 °C for 0.5 hr. To the mixture is added dropwise a 2-diazoacetate ester (1.7 eq) and the mixture is stirred at 15 °C until the reaction is complete. The mixture is concentrated under reduced pressure. The residue is added dropwise to concentrated H 2 SO 4 and stirred at 15 °C until the reaction is complete. The mixture is added to saturated NaHCO 3 solution to adjust the pH to 8 and extracted with EtOAc.
  • BF 3 .Et 2 O 0.1 eq
  • Step 2 To a solution of the protected benzofuran-3-carboxylate 2 (10 mmol, 1 eq) in MeOH (50 mL) is added Mg (5 eq) and the mixture is stirred at 15°C until the reaction is complete. Aqueous HCl (1 N, 100 mL) is added at 0 °C, and the mixture is extracted with EtOAc. The organic phase is washed with saturated brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue is purified by chromatography over silica gel (eluent petroleum ether / ethyl acetate gradient) to give the product 3.
  • Step 3 To a solution of the protected 2,3-dihydrobenzofuran-3-carboxylate 3 (1 mmol, 1 eq) cooled to 0 °C in an inert solvent such as DMF (5 mL) is added a deprotonating agent such as NaH (1.5 eq ) slowly at 0 °C, and the mixture is stirred at 0 °C for 0.5 hr. To the mixture is added dropwise the 1-fluoro-2-nitrobenzene reagent (1.2 eq) at 0 °C and the mixture is stirred at 15 °C until the reaction is complete. The mixture is quenched by addition water (50 mL) at 0°C and extracted with EtOAc.
  • a deprotonating agent such as NaH (1.5 eq ) slowly at 0 °C
  • NaH 1.5 eq
  • Step 4 A mixture of 2,3-dihydrobenzofuran-3-carboxylate 4 (1 mmol, 1 eq) and Na 2 S 2 O 4 (10 eq) in EtOH (10 mL) and H 2 O (2 mL) is stirred at 80 °C until the reaction is complete. The mixture is filtered and concentrated under reduced pressure. The residue is purified by chromatography over silica gel (eluent petroleum ether / ethyl acetate gradient) to give product 5.
  • Step 1 To a stirred mixture of arylprop-2-enoic acid 1 (40 mmol, 1 eq) and (COCl) 2 (5 eq) in DCM (70 mL) cooled on ice is added DMF (0.3 eq) and the mixture is stirred at 0°C for 1 h. The solvent is evaporated and the residue is dissolved in DCM and added to the 2-bromoaniline derivative (1.2 eq) and TEA (2 eq) at 0 °C. Then the mixture is stirred until the reaction is complete. The mixture is diluted with water and extracted with EtOAc. The organic phase is washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue is purified by chromatography over silica gel (eluent petroleum ether / ethyl acetate gradient) to give product 2.
  • DMF 0.3 eq
  • Step 2 To a stirred solution of 2 (30 mmol, 1 eq) in an inert solvent such as THF (140 mL) is added a deprotonating agent such as NaH (3 eq) at 0 °C. A benzylic halide such a paramethoxybenzyl chloride (2.5 eq) is added and the mixture is warmed to 15°C and stirred until th reaction is complete. The mixture is diluted with water and extracted with EtOAc. The organic phase is washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue is purified by chromatography over silica gel (eluent petroleum ether / ethyl acetate gradient) to give product 3.
  • a deprotonating agent such as NaH (3 eq) at 0 °C.
  • a benzylic halide such as paramethoxybenzyl chloride (2.5 eq) is added and the mixture is warmed to 15°C
  • Step 3 A mixture of 3 (6 mmol, 1 eq), K 2 CO 3 (3 eq), CH 2 Br 2 (8 eq) and a suitable catalyst such as [2-(2-aminophenyl)phenyl]-chloro-palladium;tritert-butylphosphane (0.15 eq) in DMA (50 mL) is degassed and purged with N 2 . The mixture is then stirred at 130 °C under N 2 atmosphere until the reaction is complete. The reaction mixture is diluted with water and extracted times with EtOAc. The organic phase is washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue is purified by chromatography to give product 4.
  • a suitable catalyst such as [2-(2-aminophenyl)phenyl]-chloro-palladium;tritert-butylphosphane (0.15 eq) in DMA (50 mL) is degassed and purged with N 2 .
  • Step 4 To a solution of 4 (1 mmol, 1 eq) in DCM (5 mL) is added TFA (100 eq) and TfOH (20 eq). The mixture is stirred at 50 °C until the reaction is complete. The mixture is diluted with water and extracted with EtOAc. The organic phase is washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue is purified by chromatography over silica gel (eluent petroleum ether / ethyl acetate gradient) to give product 5.
  • Step 1 To a mixture of the 2-bromobenzoic acid derivative 1 (50 mmol, 1 eq) and CuBr (0.056 eq) in ethyl 3-oxobutanoate (13.42 eq) is added NaH (2.4 eq) in one portion at 25°C under N 2 . The mixture is stirred at 80 °C until the reaction is complete. The mixture is poured into water and washed with EtOAc. The aqueous phase is acidified to pH 1 with concentrated HCI and extracted with EtOAc. The organic phase is washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. Purification of the residue by trituration or chromatography gives product 2.
  • Step 2 To a solution of benzoic acid derivative 2 (25 mmol, 1 eq) in MeOH (40 mL) is added dropwise SOCl 2 (5 eq). The mixture is stirred at 60 °C until the reaction is complete. The reaction mixture is concentrated under reduced pressure and the residue is purified by chromatography to give product 3.
  • Step 3 To a mixture of diester derivative 3 (10 mmol, 1 eq) and the 1-chloro-2-nitrobenzene reagent (1.02 eq) in THF (30 mL) is added t-BuOK (2 eq) in one portion at 25 °C under N 2 . The mixture is stirred at 25°C until the reaction is complete. The mixture is diluted with water and extracted with EtOAc. The organic phase is dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue is purified by column to give product 4.
  • Step 4 To a mixture of ester 4 (8 mmol, 1 eq) in THF (30 mL) is added LiHMDS (1.3 eq) at -78°C under N 2 . The mixture is stirred at -78 °C for 30 min. NFSI (1.02 eq) is added and the mixture is stirred until the reaction is complete. The mixture is diluted with water, extracted with EtOAc, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue is purified by chromatography to give product 5.
  • Step 5 To a mixture of the alphafluoro ester 5 (1 mmol, 1 eq) in AcOH (3 mL) is added Fe (10 eq) in one portion at 25°C under N 2 . The mixture is stirred at 60 °C until the reaction is complete. The mixture is filtered and concentrated under reduced pressure and the residue is purified by chromatography to give product 6.
  • Step 6 A solution of the 3-fluoroindolinone 6 (0.5 mmol, 1 eq) in AcOH (2 mL) and HCl (1 mL) is stirred at 100 °C for 1.5 hours. The mixture is filtered and concentrated under reduced pressure and the residue is purified by chromatography to give the product 7.
  • Step 1 To a solution of indoline-2,3-dione derivative 1 (5 mmol, 1 eq) in THF (10 mL) is added a deprotonating agent such as NaH (1.5 eq) slowly at 0 °C, and the mixture is stirred for 1 h. A solution of the heteroaryl Grignard reagent, prepared under standard conditions, is added under N 2 and the mixture is stirred at 15 °C until the reaction is complete. The mixture is diluted with water and extracted with EtOAc. The organic phase is washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue is purified by chromatography over silica gel (eluent petroleum ether / ethyl acetate gradient) to give product 2.
  • a deprotonating agent such as NaH (1.5 eq) slowly at 0 °C
  • a solution of the heteroaryl Grignard reagent prepared under standard conditions, is added under N 2 and the mixture is stir
  • Step 2 To a solution of indolin-2-one 2 (0.5 mmol, 1 eq) in DCM (5 mL) is added SOCl 2 (5 eq). The mixture is stirred at 50 °C under N 2 until the reaction is complete. The reaction is quenched with water and extracted with EtOAc. The organic phase is washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue is purified by chromatography over silica gel (eluent petroleum ether / ethyl acetate gradient) to give product 3 .
  • Step 3 To a solution of 3-chloro-indolin-2-one 3 (0.5 mmol, 1 eq) in THF cooled to 0 °C is added dropwise a trialkyl aluminum, in an inert solvent (8 eq). The mixture is warmed to 50 °C and stirred for N 2 until the reaction is complete. The reaction is quenched with water and extracted with EtOAc. The organic phase is washed with brine (10 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue is purified by chromatography to give product 4.
  • hydrocarbon group refers to a group consisting of carbon atoms and hydrogen atoms.
  • alicyclic is used in connection with cyclic groups and denotes that the corresponding cyclic group is non-aromatic.
  • alkyl refers to a monovalent saturated acyclic (i.e., non-cyclic) hydrocarbon group which may be linear or branched. Accordingly, an “alkyl” group does not comprise any carbon-to-carbon double bond or any carbon-to-carbon triple bond.
  • a "C 1-5 alkyl” denotes an alkyl group having 1 to 5 carbon atoms. Preferred exemplary alkyl groups are methyl, ethyl, propyl (e.g., n-propyl or isopropyl), or butyl (e.g., n-butyl, isobutyl, sec-butyl, or tert-butyl).
  • alkyl preferably refers to C 1-4 alkyl, more preferably to methyl or ethyl, and even more preferably to methyl.
  • alkenyl refers to a monovalent unsaturated acyclic hydrocarbon group which may be linear or branched and comprises one or more (e.g., one or two) carbon-to-carbon double bonds while it does not comprise any carbon-to-carbon triple bond.
  • C 2-5 alkenyl denotes an alkenyl group having 2 to 5 carbon atoms.
  • Preferred exemplary alkenyl groups are ethenyl, propenyl (e.g., prop-1-en-1-yl, prop-1-en-2-yl, or prop-2-en-1-yl), butenyl, butadienyl (e.g., buta-1,3-dien-1-yl or buta-1,3-dien-2-yl), pentenyl, or pentadienyl (e.g., isoprenyl).
  • alkenyl preferably refers to C 2-4 alkenyl.
  • alkynyl refers to a monovalent unsaturated acyclic hydrocarbon group which may be linear or branched and comprises one or more (e.g., one or two) carbon-to-carbon triple bonds and optionally one or more (e.g., one or two) carbon-to-carbon double bonds.
  • C 2-5 alkynyl denotes an alkynyl group having 2 to 5 carbon atoms.
  • Preferred exemplary alkynyl groups are ethynyl, propynyl (e.g., propargyl), or butynyl.
  • alkynyl preferably refers to C 2-4 alkynyl.
  • alkylene refers to an alkanediyl group, i.e. a divalent saturated acyclic hydrocarbon group which may be linear or branched.
  • a "C 1-5 alkylene” denotes an alkylene group having 1 to 5 carbon atoms, and the term “C 0-4 alkylene” indicates that a covalent bond (corresponding to the option "C 0 alkylene”) or a C 1-4 alkylene is present.
  • Preferred exemplary alkylene groups are methylene (-CH 2 -), ethylene (e.g., -CH 2 -CH 2 - or -CH(-CH 3 )-), propylene (e.g., -CH 2 -CH 2 -CH 2 -, -CH(-CH 2 -CH 3 )-, -CH 2 -CH(-CH 3 )-, or -CH(-CH 3 )-CH 2 -), or butylene (e.g., -CH 2 -CH 2 -CH 2 -CH 2 -CH 2 -).
  • alkylene preferably refers to C 1-4 alkylene (including, in particular, linear C 1-4 alkylene), more preferably to methylene or ethylene, and even more preferably to methylene.
  • alkenylene refers to an alkenediyl group, i.e. a divalent unsaturated acyclic hydrocarbon group which may be linear or branched and comprises one or more (e.g., one or two) carbon-to-carbon double bonds while it does not comprise any carbon-to-carbon triple bond.
  • a "C 2-5 alkenylene” denotes an alkenylene group having 2 to 5 carbon atoms.
  • carbocyclyl refers to a hydrocarbon ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings), wherein said ring group may be saturated, partially unsaturated (i.e., unsaturated but not aromatic) or aromatic.
  • “carbocyclyl” preferably refers to aryl, cycloalkyl or cycloalkenyl.
  • heterocyclyl refers to a ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings), wherein said ring group comprises one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from O, S and N, and the remaining ring atoms are carbon atoms, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) may optionally be oxidized, wherein one or more carbon ring atoms may optionally be oxidized (i.e., to form an oxo group), and further wherein said ring group may be saturated, partially unsaturated (i.e., unsaturated but not aromatic) or aromatic.
  • each heteroatom-containing ring comprised in said ring group may contain one or two O atoms and/or one or two S atoms (which may optionally be oxidized) and/or one, two, three or four N atoms (which may optionally be oxidized), provided that the total number of heteroatoms in the corresponding heteroatom-containing ring is 1 to 4 and that there is at least one carbon ring atom (which may optionally be oxidized) in the corresponding heteroatom-containing ring.
  • heterocyclyl preferably refers to heteroaryl, heterocycloalkyl or heterocycloalkenyl.
  • aryl refers to an aromatic hydrocarbon ring group, including monocyclic aromatic rings as well as bridged ring and/or fused ring systems containing at least one aromatic ring (e.g., ring systems composed of two or three fused rings, wherein at least one of these fused rings is aromatic; or bridged ring systems composed of two or three rings, wherein at least one of these bridged rings is aromatic).
  • aryl is a bridged and/or fused ring system which contains, besides one or more aromatic rings, at least one non-aromatic ring (e.g., a saturated ring or an unsaturated alicyclic ring), then one or more carbon ring atoms in each non-aromatic ring may optionally be oxidized (i.e., to form an oxo group).
  • non-aromatic ring e.g., a saturated ring or an unsaturated alicyclic ring
  • carbon ring atoms in each non-aromatic ring may optionally be oxidized (i.e., to form an oxo group).
  • Aryl may, e.g., refer to phenyl, naphthyl, dialinyl (i.e., 1,2-dihydronaphthyl), tetralinyl (i.e., 1,2,3,4-tetrahydronaphthyl), indanyl, indenyl (e.g., 1H-indenyl), anthracenyl, phenanthrenyl, 9H-fluorenyl, or azulenyl.
  • dialinyl i.e., 1,2-dihydronaphthyl
  • tetralinyl i.e., 1,2,3,4-tetrahydronaphthyl
  • indanyl e.g., indenyl (e.g., 1H-indenyl), anthracenyl, phenanthrenyl, 9H-fluorenyl, or azulenyl.
  • an "aryl” preferably has 6 to 14 ring atoms, more preferably 6 to 10 ring atoms, even more preferably refers to phenyl or naphthyl, and most preferably refers to phenyl.
  • heteroaryl refers to an aromatic ring group, including monocyclic aromatic rings as well as bridged ring and/or fused ring systems containing at least one aromatic ring (e.g., ring systems composed of two or three fused rings, wherein at least one of these fused rings is aromatic; or bridged ring systems composed of two or three rings, wherein at least one of these bridged rings is aromatic), wherein said aromatic ring group comprises one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from O, S and N, and the remaining ring atoms are carbon atoms, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) may optionally be oxidized, and further wherein one or more carbon ring atoms may optionally be oxidized (i.e., to form an oxo group).
  • aromatic ring group comprises one or more (such as, e.g., one, two,
  • each heteroatom-containing ring comprised in said aromatic ring group may contain one or two O atoms and/or one or two S atoms (which may optionally be oxidized) and/or one, two, three or four N atoms (which may optionally be oxidized), provided that the total number of heteroatoms in the corresponding heteroatom-containing ring is 1 to 4 and that there is at least one carbon ring atom (which may optionally be oxidized) in the corresponding heteroatom-containing ring.
  • Heteroaryl may, e.g., refer to thienyl (i.e., thiophenyl), benzo[b]thienyl, naphtho[2,3-b]thienyl, thianthrenyl, furyl (i.e., furanyl), benzofuranyl, isobenzofuranyl, chromanyl, chromenyl (e.g., 2H-1-benzopyranyl or 4H-1-benzopyranyl), isochromenyl (e.g., 1H-2-benzopyranyl), chromonyl, xanthenyl, phenoxathiinyl, pyrrolyl (e.g., 1H-pyrrolyl), imidazolyl, pyrazolyl, pyridyl (i.e., pyridinyl; e.g., 2-pyridyl, 3-pyridyl, or 4-pyridyl), pyrazin
  • heteroaryl preferably refers to a 5 to 14 membered (more preferably 5 to 10 membered) monocyclic ring or fused ring system comprising one or more (e.g., one, two, three or four) ring heteroatoms independently selected from O, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized; even more preferably, a “heteroaryl” refers to a 5 or 6 membered monocyclic ring comprising one or more (e.g., one, two or three) ring heteroatoms independently selected from O, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized;
  • heteroaryl examples include pyridinyl (e.g., 2-pyridyl, 3-pyridyl, or 4-pyridyl), imidazolyl, thiazolyl, 1H-tetrazolyl, 2H-tetrazolyl, thienyl (i.e., thiophenyl), or pyrimidinyl.
  • cycloalkyl refers to a saturated hydrocarbon ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings; such as, e.g., a fused ring system composed of two or three fused rings).
  • Cycloalkyl may, e.g., refer to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, decalinyl (i.e., decahydronaphthyl), or adamantyl.
  • cycloalkyl preferably refers to a C 3-11 cycloalkyl, and more preferably refers to a C 3-7 cycloalkyl.
  • a particularly preferred "cycloalkyl” is a monocyclic saturated hydrocarbon ring having 3 to 7 ring members.
  • particularly preferred examples of a “cycloalkyl” include cyclohexyl or cyclopropyl, particularly cyclohexyl.
  • heterocycloalkyl refers to a saturated ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings; such as, e.g., a fused ring system composed of two or three fused rings), wherein said ring group contains one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from O, S and N, and the remaining ring atoms are carbon atoms, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) may optionally be oxidized, and further wherein one or more carbon ring atoms may optionally be oxidized (i.e., to form an oxo group).
  • ring group contains one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from O
  • each heteroatom-containing ring comprised in said saturated ring group may contain one or two O atoms and/or one or two S atoms (which may optionally be oxidized) and/or one, two, three or four N atoms (which may optionally be oxidized), provided that the total number of heteroatoms in the corresponding heteroatom-containing ring is 1 to 4 and that there is at least one carbon ring atom (which may optionally be oxidized) in the corresponding heteroatom-containing ring.
  • Heterocycloalkyl may, e.g., refer to aziridinyl, azetidinyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, piperidinyl, piperazinyl, azepanyl, diazepanyl (e.g., 1,4-diazepanyl), oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, morpholinyl (e.g., morpholin-4-yl), thiomorpholinyl (e.g., thiomorpholin-4-yl), oxazepanyl, oxiranyl, oxetanyl, tetrahydrofuranyl, 1,3-dioxolanyl, tetrahydropyranyl, 1,4-dioxanyl, oxepanyl, thiiran
  • heterocycloalkyl preferably refers to a 3 to 11 membered saturated ring group, which is a monocyclic ring or a fused ring system (e.g., a fused ring system composed of two fused rings), wherein said ring group contains one or more (e.g., one, two, three, or four) ring heteroatoms independently selected from O, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized; more preferably, "heterocycloalkyl” refers to a 5 to 7 membered saturated monocyclic ring group containing one or more (e.g., one, two, or three) ring heteroatoms independently selected from O, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring
  • heterocycloalkyl examples include tetrahydropyranyl, piperidinyl, piperazinyl, morpholinyl, pyrrolidinyl, or tetrahydrofuranyl.
  • cycloalkenyl refers to an unsaturated alicyclic (non-aromatic) hydrocarbon ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings; such as, e.g., a fused ring system composed of two or three fused rings), wherein said hydrocarbon ring group comprises one or more (e.g., one or two) carbon-to-carbon double bonds and does not comprise any carbon-to-carbon triple bond.
  • Cycloalkenyl may, e.g., refer to cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptenyl, or cycloheptadienyl.
  • cycloalkenyl preferably refers to a C 3-11 cycloalkenyl, and more preferably refers to a C 3-7 cycloalkenyl.
  • a particularly preferred "cycloalkenyl” is a monocyclic unsaturated alicyclic hydrocarbon ring having 3 to 7 ring members and containing one or more (e.g., one or two; preferably one) carbon-to-carbon double bonds.
  • heterocycloalkenyl refers to an unsaturated alicyclic (non-aromatic) ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings; such as, e.g., a fused ring system composed of two or three fused rings), wherein said ring group contains one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from O, S and N, and the remaining ring atoms are carbon atoms, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) may optionally be oxidized, wherein one or more carbon ring atoms may optionally be oxidized (i.e., to form an oxo group), and further wherein said ring group comprises at least one double bond between
  • each heteroatom-containing ring comprised in said unsaturated alicyclic ring group may contain one or two O atoms and/or one or two S atoms (which may optionally be oxidized) and/or one, two, three or four N atoms (which may optionally be oxidized), provided that the total number of heteroatoms in the corresponding heteroatom-containing ring is 1 to 4 and that there is at least one carbon ring atom (which may optionally be oxidized) in the corresponding heteroatom-containing ring.
  • Heterocycloalkenyl may, e.g., refer to imidazolinyl (e.g., 2-imidazolinyl (i.e., 4,5-dihydro-1H-imidazolyl), 3-imidazolinyl, or 4-imidazolinyl), tetrahydropyridinyl (e.g., 1,2,3,6-tetrahydropyridinyl), dihydropyridinyl (e.g., 1,2-dihydropyridinyl or 2,3-dihydropyridinyl), pyranyl (e.g., 2H-pyranyl or 4H-pyranyl), thiopyranyl (e.g., 2H-thiopyranyl or 4H-thiopyranyl), dihydropyranyl, dihydrofuranyl, dihydropyrazolyl, dihydropyrazinyl, dihydroisoindolyl, oct
  • heterocycloalkenyl preferably refers to a 3 to 11 membered unsaturated alicyclic ring group, which is a monocyclic ring or a fused ring system (e.g., a fused ring system composed of two fused rings), wherein said ring group contains one or more (e.g., one, two, three, or four) ring heteroatoms independently selected from O, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, wherein one or more carbon ring atoms are optionally oxidized, and wherein said ring group comprises at least one double bond between adjacent ring atoms and does not comprise any triple bond between adjacent ring atoms; more preferably, "heterocycloalkenyl” refers to a 5 to 7 membered monocyclic unsaturated non-aromatic ring group containing one or more (e.
  • halogen refers to fluoro (-F), chloro (-CI), bromo (-Br), or iodo (-I).
  • haloalkyl refers to an alkyl group substituted with one or more (preferably 1 to 6, more preferably 1 to 3) halogen atoms which are selected independently from fluoro, chloro, bromo and iodo, and are preferably all fluoro atoms. It will be understood that the maximum number of halogen atoms is limited by the number of available attachment sites and, thus, depends on the number of carbon atoms comprised in the alkyl moiety of the haloalkyl group.
  • Haloalkyl may, e.g., refer to -CF 3 , -CHF 2 , -CH 2 F, -CF 2 -CH 3 , -CH 2 -CF 3 , -CH 2 -CHF 2 , -CH 2 -CF 2 -CH 3 , -CH 2 -CF 2 -CF 3 , or -CH(CF 3 ) 2 .
  • a particularly preferred "haloalkyl" group is -CF 3 .
  • the terms “optional”, “optionally” and “may” denote that the indicated feature may be present but can also be absent.
  • the present invention specifically relates to both possibilities, i.e., that the corresponding feature is present or, alternatively, that the corresponding feature is absent.
  • the expression “X is optionally substituted with Y" (or “X may be substituted with Y”) means that X is either substituted with Y or is unsubstituted.
  • a component of a composition is indicated to be “optional”
  • the invention specifically relates to both possibilities, i.e., that the corresponding component is present (contained in the composition) or that the corresponding component is absent from the composition.
  • substituents such as, e.g., one, two, three or four substituents. It will be understood that the maximum number of substituents is limited by the number of attachment sites available on the substituted moiety.
  • the "optionally substituted" groups referred to in this specification carry preferably not more than two substituents and may, in particular, carry only one substituent.
  • the optional substituents are absent, i.e. that the corresponding groups are unsubstituted.
  • substituent groups comprised in the compounds of the present invention may be attached to the remainder of the respective compound via a number of different positions of the corresponding specific substituent group. Unless defined otherwise, the preferred attachment positions for the various specific substituent groups are as illustrated in the examples.
  • compositions comprising “a” compound of formula (I) can be interpreted as referring to a composition comprising "one or more” compounds of formula (I).
  • the term "about” preferably refers to ⁇ 10% of the indicated numerical value, more preferably to ⁇ 5% of the indicated numerical value, and in particular to the exact numerical value indicated. If the term “about” is used in connection with the endpoints of a range, it preferably refers to the range from the lower endpoint -10% of its indicated numerical value to the upper endpoint +10% of its indicated numerical value, more preferably to the range from of the lower endpoint -5% to the upper endpoint +5%, and even more preferably to the range defined by the exact numerical values of the lower endpoint and the upper endpoint.
  • the term “about” is used in connection with the endpoint of an open-ended range, it preferably refers to the corresponding range starting from the lower endpoint -10% or from the upper endpoint +10%, more preferably to the range starting from the lower endpoint -5% or from the upper endpoint +5%, and even more preferably to the open-ended range defined by the exact numerical value of the corresponding endpoint. If the term “about” is used in connection with a parameter that is quantified in integers, such as the number of CGG repeats in the FMR1 gene or the FMR2 gene, then the numbers corresponding to ⁇ 10% or ⁇ 5% of the indicated numerical value are to be rounded to the nearest integer (using the tie-breaking rule "round half up").
  • the term “comprising” (or “comprise”, “comprises”, “contain”, “contains”, or “containing”), unless explicitly indicated otherwise or contradicted by context, has the meaning of “containing, inter alia”, i.e., “containing, among further optional elements, ". In addition thereto, this term also includes the narrower meanings of “consisting essentially of” and “consisting of”.
  • a comprising B and C has the meaning of "A containing, inter alia, B and C", wherein A may contain further optional elements (e.g., "A containing B, C and D" would also be encompassed), but this term also includes the meaning of "A consisting essentially of B and C” and the meaning of "A consisting of B and C" (i.e., no other components than B and C are comprised in A).
  • the scope of the present invention embraces all pharmaceutically acceptable salt forms of the compounds of formula (I) or (Ia) which may be formed, e.g., by protonation of an atom carrying an electron lone pair which is susceptible to protonation, such as an amino group, with an inorganic or organic acid, or as a salt of an acid group (such as a carboxylic acid group) with a physiologically acceptable cation.
  • Exemplary base addition salts comprise, for example: alkali metal salts such as sodium or potassium salts; alkaline earth metal salts such as calcium or magnesium salts; zinc salts; ammonium salts; aliphatic amine salts such as trimethylamine, triethylamine, dicyclohexylamine, ethanolamine, diethanolamine, triethanolamine, procaine salts, meglumine salts, ethylenediamine salts, or choline salts; aralkyl amine salts such as N,N-dibenzylethylenediamine salts, benzathine salts, benethamine salts; heterocyclic aromatic amine salts such as pyridine salts, picoline salts, quinoline salts or isoquinoline salts; quaternary ammonium salts such as tetramethylammonium salts, tetraethylammonium salts, benzyltrimethylammonium salts, benzyltriethylam
  • Exemplary acid addition salts comprise, for example: mineral acid salts such as hydrochloride, hydrobromide, hydroiodide, sulfate salts (such as, e.g., sulfate or hydrogensulfate salts), nitrate salts, phosphate salts (such as, e.g., phosphate, hydrogenphosphate, or dihydrogenphosphate salts), carbonate salts, hydrogencarbonate salts, perchlorate salts, borate salts, or thiocyanate salts; organic acid salts such as acetate, propionate, butyrate, pentanoate, hexanoate, heptanoate, octanoate, cyclopentanepropionate, decanoate, undecanoate, oleate, stearate, lactate, maleate, oxalate, fumarate, tartrate, malate, citrate, succinate, adipate, gluconate, glycolate, nic
  • Preferred pharmaceutically acceptable salts of the compounds of formula (I) or (Ia) include a hydrochloride salt, a hydrobromide salt, a mesylate salt, a sulfate salt, a tartrate salt, a fumarate salt, an acetate salt, a citrate salt, and a phosphate salt.
  • a particularly preferred pharmaceutically acceptable salt of the compound of formula (I) or (Ia) is a hydrochloride salt.
  • the compound of formula (I) or (Ia), including any one of the specific compounds of formula (I) or (Ia) described herein, is in the form of a hydrochloride salt, a hydrobromide salt, a mesylate salt, a sulfate salt, a tartrate salt, a fumarate salt, an acetate salt, a citrate salt, or a phosphate salt, and it is particularly preferred that the compound of formula (I) or (Ia) is in the form of a hydrochloride salt.
  • the present invention also specifically relates to the compound of formula (I) or (Ia), including any one of the specific compounds of formula (I) or (Ia) described herein (including in the examples section), in non-salt form.
  • the compound of formula (I) or (Ia) is employed in non-salt form.
  • the scope of the invention embraces the compounds of formula (I) or (Ia) in any solvated form, including, e.g., solvates with water (i.e., as a hydrate) or solvates with organic solvents such as, e.g., methanol, ethanol, isopropanol, acetic acid, ethyl acetate, ethanolamine, DMSO, or acetonitrile. All physical forms, including any amorphous or crystalline forms (i.e., polymorphs), of the compounds of formula (I) or (Ia) are also encompassed within the scope of the invention. It is to be understood that such solvates and physical forms of pharmaceutically acceptable salts of the compounds of the formula (I) or (Ia) are likewise embraced by the invention.
  • the compounds of formula (I) or (Ia) may exist in the form of different isomers, in particular stereoisomers (including, e.g., geometric isomers (or cis/trans isomers), atropisomers, enantiomers and diastereomers) or tautomers (including, in particular, prototropic tautomers, such as keto/enol tautomers or thione/thiol tautomers). All such isomers of the compounds of formula (I) or (Ia) are contemplated as being part of the present invention, either in admixture or in pure or substantially pure form.
  • the invention embraces the isolated optical isomers of the compounds according to the invention as well as any mixtures thereof (including, in particular, racemic mixtures/racemates).
  • the racemates can be resolved by physical methods, such as, e.g., fractional crystallization, separation or crystallization of diastereomeric derivatives, or separation by chiral column chromatography.
  • the individual optical isomers can also be obtained from the racemates via salt formation with an optically active acid followed by crystallization.
  • the present invention further encompasses any tautomers of the compounds of formula (I) or (Ia). It will be understood that some compounds may exhibit tautomerism. In such cases, the formulae provided herein expressly depict only one of the possible tautomeric forms.
  • the formulae and chemical names as provided herein are intended to encompass any tautomeric form of the corresponding compound and not to be limited merely to the specific tautomeric form depicted by the drawing or identified by the name of the compound.
  • the compounds of formula (I) or (Ia) may have the (S)-configuration or the (R)-configuration at the carbon atom carrying the group R 1 , as illustrated in the following (see also Examples 2 and 3):
  • the present invention specifically relates to an optical isomer (or optically active isomer) of the compound of formula (I) or (Ia), wherein the carbon atom carrying R 1 is in (S)-configuration, and further specifically relates to an optical isomer (or optically active isomer) of the compound of formula (I) or (Ia), wherein the carbon atom carrying R 1 is in (R)-configuration.
  • the invention also relates to any mixtures of such isomers, including a corresponding racemic mixture.
  • the scope of the invention further embraces compounds of formula (I) or (Ia), in which one or more atoms are replaced by a specific isotope of the corresponding atom.
  • the invention encompasses compounds of formula (I) or (Ia), in which one or more hydrogen atoms (or, e.g., all hydrogen atoms) are replaced by deuterium atoms (i.e., 2 H; also referred to as "D").
  • the invention also embraces compounds of formula (I) or (Ia) which are enriched in deuterium.
  • Naturally occurring hydrogen is an isotopic mixture comprising about 99.98 mol-% hydrogen-1 ( 1 H) and about 0.0156 mol-% deuterium ( 2 H or D).
  • the content of deuterium in one or more hydrogen positions in the compounds of formula (I) or (Ia) can be increased using deuteration techniques known in the art.
  • a compound of formula (I) or (Ia), or a reactant or precursor to be used in the synthesis of the compound of formula (I) or (Ia) can be subjected to an H/D exchange reaction using, e.g., heavy water (D 2 O).
  • deuteration techniques are described in: Atzrodt J et al., Bioorg Med Chem, 2012, 20(18): 5658-67 ; William JS et al., Journal of Labelled Compounds and Radiopharmaceuticals, 2010, 53(11-12): 635-44 ; Modvig A et al., J Org Chem, 2014, 79: 5861-8 .
  • the content of deuterium can be determined, e.g., using mass spectrometry or NMR spectroscopy. Unless specifically indicated otherwise, it is preferred that the compound of formula (I) or (Ia) is not enriched in deuterium. Accordingly, the presence of naturally occurring hydrogen atoms or 1 H hydrogen atoms in the compounds of formula (I) or (Ia) is preferred.
  • the present invention also embraces compounds of formula (I) or (Ia), in which one or more atoms are replaced by a positron-emitting isotope of the corresponding atom, such as, e.g., 18 F, 11 C, 13 N, 15 O, 76 Br, 77 Br, 120 I and/or 124 I.
  • a positron-emitting isotope of the corresponding atom such as, e.g., 18 F, 11 C, 13 N, 15 O, 76 Br, 77 Br, 120 I and/or 124 I.
  • Such compounds can be used as tracers, trackers or imaging probes in positron emission tomography (PET).
  • the invention thus includes (i) compounds of formula (I) or (Ia), in which one or more fluorine atoms (or, e.g., all fluorine atoms) are replaced by 18 F atoms, (ii) compounds of formula (I) or (Ia), in which one or more carbon atoms (or, e.g., all carbon atoms) are replaced by 11 C atoms, (iii) compounds of formula (I) or (Ia), in which one or more nitrogen atoms (or, e.g., all nitrogen atoms) are replaced by 13 N atoms, (iv) compounds of formula (I) or (Ia), in which one or more oxygen atoms (or, e.g., all oxygen atoms) are replaced by 15 O atoms, (v) compounds of formula (I) or (Ia), in which one or more bromine atoms (or, e.g., all bromine atoms) are replaced by 76 Br atoms, (vi) compounds
  • the compounds of the present invention may be administered as compounds per se or may be formulated as medicaments/pharmaceutical compositions.
  • the medicaments/pharmaceutical compositions may optionally comprise one or more pharmaceutically acceptable excipients, such as carriers, diluents, fillers, disintegrants, lubricating agents, binders, colorants, pigments, stabilizers, preservatives, antioxidants, and/or solubility enhancers.
  • the pharmaceutical compositions may comprise one or more solubility enhancers, such as, e.g., poly(ethylene glycol), including poly(ethylene glycol) having a molecular weight in the range of about 200 to about 5,000 Da (e.g., PEG 200, PEG 300, PEG 400, or PEG 600), ethylene glycol, propylene glycol, glycerol, a non-ionic surfactant, tyloxapol, polysorbate 80, macrogol-15-hydroxystearate (e.g., Kolliphor® HS 15, CAS 70142-34-6), a phospholipid, lecithin, dimyristoyl phosphatidylcholine, dipalmitoyl phosphatidylcholine, distearoyl phosphatidylcholine, a cyclodextrin, ⁇ -cydodextrin, ⁇ -cydodextrin, ⁇ -cyclodextrin, hydroxyethyl- ⁇ -cycl
  • the pharmaceutical compositions may also comprise one or more preservatives, particularly one or more antimicrobial preservatives, such as, e.g., benzyl alcohol, chlorobutanol, 2-ethoxyethanol, m-cresol, chlorocresol (e.g., 2-chloro-3-methyl-phenol or 4-chloro-3-methylphenol), benzalkonium chloride, benzethonium chloride, benzoic acid (or a pharmaceutically acceptable salt thereof), sorbic acid (or a pharmaceutically acceptable salt thereof), chlorhexidine, thimerosal, or any combination thereof.
  • preservatives particularly one or more antimicrobial preservatives, such as, e.g., benzyl alcohol, chlorobutanol, 2-ethoxyethanol, m-cresol, chlorocresol (e.g., 2-chloro-3-methyl-phenol or 4-chloro-3-methylphenol), benzalkonium chloride, benzethonium chloride, benzoic acid (
  • compositions can be formulated by techniques known to the person skilled in the art, such as the techniques published in " Remington: The Science and Practice of Pharmacy", Pharmaceutical Press, 22nd editi on.
  • the pharmaceutical compositions can be formulated as dosage forms for oral, parenteral, such as intramuscular, intravenous, subcutaneous, intradermal, intraarterial, intracardial, rectal, nasal, topical, aerosol or vaginal administration.
  • Dosage forms for oral administration include coated and uncoated tablets, soft gelatin capsules, hard gelatin capsules, lozenges, troches, solutions, emulsions, suspensions, syrups, elixirs, powders and granules for reconstitution, dispersible powders and granules, medicated gums, chewing tablets and effervescent tablets.
  • Dosage forms for parenteral administration include solutions, emulsions, suspensions, dispersions and powders and granules for reconstitution. Emulsions are a preferred dosage form for parenteral administration.
  • Dosage forms for rectal and vaginal administration include suppositories and ovula.
  • Dosage forms for nasal administration can be administered via inhalation and insufflation, for example by a metered inhaler.
  • Dosage forms for topical administration include creams, gels, ointments, salves, patches and transdermal delivery systems.
  • the compounds of formula (I) or (Ia), or the above described pharmaceutical compositions comprising a compound of formula (I) or (Ia), may be administered to a subject by any convenient route of administration, whether systemically/peripherally or at the site of desired action, including but not limited to one or more of: oral (e.g., as a tablet, capsule, or as an ingestible solution), topical (e.g., transdermal, intranasal, ocular, buccal, and sublingual), parenteral (e.g., using injection techniques or infusion techniques, and including, for example, by injection, e.g., subcutaneous, intradermal, intramuscular, intravenous, intraarterial, intracardiac, intrathecal, intraspinal, intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, subcuticular, intraarticular, subarachnoid, or intrasternal by, e.g., implant of a depot, for example, subcutaneously
  • examples of such administration include one or more of: intravenously, intraarterially, intraperitoneally, intrathecally, intraventricularly, intraurethrally, intrasternally, intracardially, intracranially, intramuscularly or subcutaneously administering the compounds or pharmaceutical compositions, and/or by using infusion techniques.
  • parenteral administration the compounds are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood.
  • the aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary.
  • the preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well known to those skilled in the art.
  • said compounds or pharmaceutical compositions are administered orally, e.g., in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavoring or coloring agents, for immediate-, delayed-, modified-, sustained-, pulsed- or controlled-release applications.
  • the tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycolate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxypropylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included. Solid compositions of a similar type may also be employed as fillers in gelatin capsules.
  • excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine
  • disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glyco
  • Preferred excipients in this regard include lactose, starch, a cellulose, or high molecular weight polyethylene glycols.
  • the agent may be combined with various sweetening or flavoring agents, coloring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
  • the compounds or pharmaceutical compositions are preferably administered by oral ingestion, particularly by swallowing.
  • the compounds or pharmaceutical compositions can thus be administered to pass through the mouth into the gastrointestinal tract, which can also be referred to as "oral-gastrointestinal" administration.
  • said compounds or pharmaceutical compositions can be administered in the form of a suppository or pessary, or may be applied topically in the form of a gel, hydrogel, lotion, solution, cream, ointment or dusting powder.
  • the compounds of the present invention may also be dermally or transdermally administered, for example, by the use of a skin patch.
  • sustained-release compositions include semi-permeable polymer matrices in the form of shaped articles, e.g., films, or microcapsules.
  • Sustained-release matrices include, e.g., polylactides, copolymers of L-glutamic acid and gamma-ethyl-L-glutamate, poly(2-hydroxyethyl methacrylate), ethylene vinyl acetate, or poly-D-(-)-3-hydroxybutyric acid.
  • Sustained-release pharmaceutical compositions also include liposomally entrapped compounds. The present invention thus also relates to liposomes containing a compound of the invention.
  • Said compounds or pharmaceutical compositions may also be administered by the pulmonary route, the rectal route, or the ocular route.
  • they can be formulated as micronized suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzalkonium chloride.
  • they may be formulated in an ointment such as petrolatum.
  • dry powder formulations of the compounds of formula (I) for pulmonary administration may be prepared by spray drying under conditions which result in a substantially amorphous glassy or a substantially crystalline bioactive powder. Accordingly, dry powders of the compounds of the present invention can be made according to an emulsification/spray drying process.
  • said compounds or pharmaceutical compositions can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, emulsifying wax and water.
  • they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, 2-octyldodecanol, benzyl alcohol and water.
  • the present invention thus relates to the compounds or the pharmaceutical compositions provided herein, wherein the corresponding compound or pharmaceutical composition is to be administered by any one of: an oral route; topical route, including by transdermal, intranasal, ocular, buccal, or sublingual route; parenteral route using injection techniques or infusion techniques, including by subcutaneous, intradermal, intramuscular, intravenous, intraarterial, intracardiac, intrathecal, intraspinal, intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, subcuticular, intraarticular, subarachnoid, intrasternal, intraventricular, intraurethral, or intracranial route; pulmonary route, including by inhalation or insufflation therapy; gastrointestinal route; intrauterine route; intraocular route; subcutaneous route; ophthalmic route, including by intravitreal, or intracameral route; rectal route; or vaginal route.
  • a preferred route of administration is oral administration.
  • a physician will determine the actual dosage which will be most suitable for an individual subject.
  • the specific dose level and frequency of dosage for any particular individual subject may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual subject undergoing therapy.
  • a proposed, yet non-limiting dose of the compounds according to the invention for oral administration to a human may be 0.05 to 2000 mg, preferably 0.1 mg to 1000 mg, of the active ingredient per unit dose.
  • the unit dose may be administered, e.g., 1 to 3 times per day.
  • the unit dose may also be administered 1 to 7 times per week, e.g., with not more than one administration per day. It will be appreciated that it may be necessary to make routine variations to the dosage depending on the age and weight of the patient/subject as well as the severity of the condition to be treated. The precise dose and also the route of administration will ultimately be at the discretion of the attendant physician or veterinarian.
  • the compound of formula (I) or (Ia), or a pharmaceutical composition comprising the compound of formula (I) or (Ia), can be administered in monotherapy (e.g., without concomitantly administering any further therapeutic agents, or without concomitantly administering any further therapeutic agents against the same disease that is to be treated or prevented with the compound of formula (I) or (Ia)).
  • monotherapy e.g., without concomitantly administering any further therapeutic agents, or without concomitantly administering any further therapeutic agents against the same disease that is to be treated or prevented with the compound of formula (I) or (Ia)
  • the present invention relates to the compound of formula (I) or (Ia), or a corresponding pharmaceutical composition, for use in the monotherapeutic treatment or prevention of a fragile X associated disorder (such as fragile X syndrome).
  • the invention relates to the monotherapeutic administration of the compound of formula (I) or (Ia), or a corresponding pharmaceutical composition, without concomitantly administering any further therapeutic agents against a fragile X associated disorder (or any further therapeutic agents against fragile X syndrome).
  • the compound of formula (I) or (Ia), or a pharmaceutical composition comprising the compound of formula (I) or (Ia), can also be administered in combination with one or more further therapeutic agents. If the compound of formula (I) or (Ia) is used in combination with a second therapeutic agent active against the same disease or condition, the dose of each compound may differ from that when the corresponding compound is used alone, in particular, a lower dose of each compound may be used.
  • the combination of the compound of formula (I) or (Ia) with one or more further therapeutic agents may comprise the simultaneous/concomitant administration of the compound of formula (I) or (Ia) and the further therapeutic agent(s) (either in a single pharmaceutical formulation or in separate pharmaceutical formulations), or the sequential/separate administration of the compound of formula (I) or (Ia) and the further therapeutic agent(s). If administration is sequential, either the compound of formula (I) or (Ia) according to the invention or the one or more further therapeutic agents may be administered first. If administration is simultaneous, the one or more further therapeutic agents may be included in the same pharmaceutical formulation as the compound of formula (I) or (Ia), or they may be administered in two or more different (separate) pharmaceutical formulations. Such different pharmaceutical formulations may be administered via the same route or via different routes of administration.
  • the subject or patient to be treated in accordance with the present invention may be an animal (e.g., a non-human animal).
  • the subject/patient is a mammal.
  • the subject/patient is a human (e.g., a male human or a female human) or a non-human mammal (such as, e.g., a guinea pig, a hamster, a rat, a mouse, a rabbit, a dog, a cat, a horse, a monkey, an ape, a marmoset, a baboon, a gorilla, a chimpanzee, an orangutan, a gibbon, a sheep, cattle, or a pig).
  • the subject/patient to be treated in accordance with the invention is a human.
  • Treatment of a disease or disorder, as used herein, is well known in the art.
  • Treatment of a disease or disorder implies that the corresponding disease or disorder is suspected or has been diagnosed in a patient/subject.
  • a patient/subject suspected of suffering from a disease or disorder typically shows specific clinical and/or pathological symptoms which a skilled person can easily attribute to a specific pathological condition (i.e., diagnose a disease or disorder).
  • the "treatment" of a disease or disorder may, for example, lead to a halt in the progression of the disease or disorder (e.g., no deterioration of symptoms) or a delay in the progression of the disease or disorder (in case the halt in progression is of a transient nature only).
  • the "treatment” of a disease or disorder may also lead to a partial response (e.g., amelioration of symptoms) or complete response (e.g., disappearance of symptoms) of the subject/patient suffering from the disease or disorder.
  • the "treatment” of a disease or disorder may also refer to an amelioration of the disease or disorder, which may, e.g., lead to a halt in the progression of the disease or disorder or a delay in the progression of the disease or disorder.
  • Such a partial or complete response may be followed by a relapse.
  • a subject/patient may experience a broad range of responses to a treatment (such as the exemplary responses as described herein above).
  • the treatment of a disease or disorder may, inter alia , comprise curative treatment (preferably leading to a complete response and eventually to healing of the disease or disorder) or palliative treatment (including symptomatic relief).
  • prevention of a disease or disorder is also well known in the art.
  • a patient/subject suspected of being prone to suffer from a disease or disorder may particularly benefit from a prevention of the disease or disorder.
  • the subject/patient may have a susceptibility or predisposition for a disease or disorder, including but not limited to hereditary predisposition.
  • Such a predisposition can be determined by standard methods or assays, using, e.g., genetic markers or phenotypic indicators.
  • a disease or disorder to be prevented in accordance with the present invention has not been diagnosed or cannot be diagnosed in the patient/subject (for example, the patient/subject does not show any clinical or pathological symptoms).
  • prevention comprises the use of a compound of the present invention before any clinical and/or pathological symptoms are diagnosed or determined or can be diagnosed or determined by the attending physician.
  • the present invention specifically relates to each and every combination of features and embodiments described herein, including any combination of general and/or preferred features/embodiments.
  • the invention specifically relates to each combination of meanings (including general and/or preferred meanings) for the various groups and variables comprised in formula (I) or (Ia).
  • the present invention particularly relates to the following items:
  • the compounds of formula (I) or (Ia) described in this section are defined by their chemical formulae and their corresponding chemical names.
  • the present invention relates to both the compound defined by the chemical formula and the compound defined by the chemical name, and particularly relates to the compound defined by the chemical formula.
  • Step 3 Preparation of ethyl 2-(5-chloro-2-methoxyphenyl)-2-(2-nitro-4-(trifluoromethyl)phenyl) acetate.
  • Step 4 Preparation of ethyl 2-(5-chloro-2-methoxyphenyl)-2-(2-nitro-4-(trifluoromethyl)phenyl) propanoate.
  • Step 1 Preparation of ethyl 2-(5-chloro-2-methoxyphenyl)-2-(2-nitro-4-(trifluoromethyl)phenyl) butanoate.
  • Example 6 was prepared following the procedure described in example 5, using propyl iodide in step 1, to give 3-(5-chloro-2-methoxyphenyl)-3-propyl-6-(trifluoromethyl)indolin-2-one (27 mg, 25.6% yield, 95% purity) as a pale yellow solid.
  • Example 7 was prepared following the procedure described in example 5, using benzyl bromide in step 1, to give 3-benzyl-3-(5-chloro-2-methoxyphenyl)-6-(trifluoromethyl)indolin-2-one (106 mg, 38.7% yield, 98.3% purity) a white solid.
  • LCMS 432.1 (M+H + ) 1 H NMR (400 MHz, CDCl 3 ): 7.678 (1H, d), 7.256 (1H, dd), 7.105, (2H, m), 7.036 (2H, m), 6.744 (4H, m), 3.505 (2H, m), 3.435 (3H, s).
  • Example 8 was prepared following the procedure described in example 5, using bromoacetonitrile in step 1, to give 2-(3-(5-chloro-2-methoxyphenyl)-2-oxo-6-(trifluoromethyl)indolin-3-yl)acetonitrile (127 mg, 33.8% yield, 93% purity) as a white solid.
  • Example 9 was prepared following the procedure described in example 5, using allyl bromide in step 1, to give 3-allyl-3-(5-chloro-2-methoxyphenyl)-6-(trifluoromethyl)indolin-2-one (0.6 g, 28.7% yield, 95.8% purity) as a white solid.
  • Step 1 Preparation of ethyl 5-chlorobenzofuran-3-carboxylate.
  • Step 2 Preparation of methyl 5-chloro-2,3-dihydrobenzofuran-3-carboxylate.
  • Step 3 Preparation of methyl 5-chloro-3-(2-nitro-4-(trifluoromethyl)phenyl)-2,3-dihydrobenzofuran-3-carboxylate.
  • reaction mixture was quenched by addition water (50 mL) at 0°C, and the mixture was extracted twice with EtOAc (50 mL). The combined organic layers were washed with saturated brine (100 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue was combined with another batch and purified by column chromatography (SiO 2 , petroleum ether / ethyl acetate 0:1 to 20:1 gradient) to give 5-chloro-3-(2-nitro-4-(trifluoromethyl)phenyl)-2,3- dihydrobenzofuran-3-carboxylate as a yellow solid.
  • Step 4 Preparation of 5-chloro-6'-(trifluoromethyl)-2H-spiro[benzofuran-3,3'-indolin]-2'-one.
  • reaction mixture was filtered and concentrated under reduced pressure and the residue was purified by column chromatography (SiO 2 , petroleum ether / ethyl acetate, 10:1 to 3:1 gradient) to give 5-chloro-6'-(trifluoromethyl)-2H-spiro[benzofuran-3,3'-indolin]-2'-one (67 mg, 20.4% yield, 98% purity) as a white solid.
  • Example 12 7-methoxy-6'-(trifluoromethyl)-2,3-dihydrospiro[indene-1,3'-indolin]-2'-one
  • Step 1 Preparation of ethyl 2-(2-methoxyphenyl)-2-oxoacetate.
  • Step 2 Preparation of ethyl 2-(2-methoxyphenyl)prop-2-enoate.
  • Step 3 Preparation of 2-(2-methoxyphenyl)prop-2-enoic acid.
  • Step 4 N-[2-bromo-5-(trifluoromethyl)phenyl]-2-(2-methoxyphenyl)prop-2-enamide.
  • Step 5 Preparation of N-[2-bromo-5-(trifluoromethyl)phenyl]-2-(2-methoxyphenyl)-N-[(4-methoxyphenyl)methyl]prop-2-enamide.
  • Step 6 Preparation of 7-methoxy-1'-[(4-methoxyphenyl)methyl]-6'-(trifluoromethyl)spiro[indane-1,3'-indoline]-2'-one
  • the mixture was then stirred at 130 °C for 12 hr under N 2 atmosphere.
  • the reaction mixture was diluted with water (20 mL) and extracted four times with EtOAc (8 mL). The combined organic layers were washed with brine (25 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure.
  • the residue was purified by prep-HPLC (column: Xtimate C18 10 ⁇ 250mm * 50mm; mobile phase: [water(10mM NH 4 HCO 3 )-ACN]; B%: 60%-70%, 20min) to give 7-methoxy-1'-[(4-methoxyphenyl)methyl]-6'-(trifluoromethyl)spiro[indane-1,3'-indoline]-2'-one (1.3 g, 2.87 mmol, 49.7% yield) as a white solid.
  • Step 7 Preparation of 7-methoxy-6'-(trifluoromethyl)spiro[indane-1,3'-indoline]-2'-one.
  • Examples 13 and 14 4-chloro-7-methoxy-6'-(trifluoromethyl)spiro[indane-1,3'-indoline]-2'-one (Example 13) and 4,6-dichloro-7-methoxy-6'-(trifluoromethyl)spiro[indane-1,3'-indoline]-2'-one (Example 14)
  • Step 1 Preparation of 3-(2-chloro-5-methoxy-4-pyridyl)-3-hydroxy-6- (trifluoromethyl)indolin-2-one
  • Step 2 Preparation of 3-chloro-3-(2-chloro-5-methoxy-4-pyridyl)-6-(trifluoromethyl)indolin -2-one
  • Step 1 Preparation of ethyl 2-(4-pyridyl)propanoate
  • Step 2 Preparation of ethyl 2-[2-nitro-4-(trifluoromethyl)phenyl]-2-(4-pyridyl)propanoate
  • Step 1 Preparation of 3-(2-chloro-4-pyridyl)-3-hydroxy-6-(trifluoromethyl)indolin-2-one.
  • Step 2 Preparation of 3-chloro-3-(2-chloro-4-pyridyl)-6-(trifluoromethyl)indolin-2-one.
  • Step 3 Preparation of 3-(2-chloro-4-pyridyl)-3-methyl-6-(trifluoromethyl)indolin-2-one.
  • reaction mixture was quenched by addition saturated NH 4 Cl 10 mL at 0°C, and then extracted three times with EtOAc (5 mL). The combined organic layers were washed twice with saturated brine (5 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure.
  • Step 1 Preparation of ethyl 2-(5-chloro-2-methoxy-phenyl)-2-[3-nitro-5-(trifluoromethyl)-2-pyridyl]acetate.
  • Step 2 Preparation of ethyl 2-(5-chloro-2-methoxy-phenyl)-2-[3-nitro-5-(trifluoromethyl)-2-pyridyl]propanoate.
  • Step 3 Preparation of 3-(5-chloro-2-methoxy-phenyl)-3-methyl-6-(trifluoromethyl)-1H-pyrrolo[3,2-b]pyridin-2-one.
  • Step 1 Preparation of 4-chloro-2-(2-ethoxy-2-oxo-ethyl)benzoic acid.
  • Step 2 Preparation of methyl 4-chloro-2-(2-methoxy-2-oxo-ethyl)benzoate.
  • Step 3 Preparation of methyl 4-chloro-2-[2-methoxy-1-[2-nitro-4-(trifluoromethyl)phenyl]-2-oxo-ethyl]benzoate.
  • Step 4 Preparation of methyl 4-chloro-2-[1-fluoro-2-methoxy-1-[2-nitro-4-(trifluoromethyl)phenyl]-2-oxo-ethyl]benzoate.
  • Step 5 Preparation of methyl 4-chloro-2-[3-fluoro-2-oxo-6-(trifluoromethyl)indolin-3-yl] benzoate.
  • Step 6 Preparation of 6'-chloro-6-(trifluoromethyl)-3'H-spiro[indoline-3,1'-isobenzofuran]-2,3'-dione.
  • Step 1 Preparation of 4-(difluoromethyl)-1-fluoro-2-nitro-benzene.
  • Step 2 Preparation of ethyl 2-(5-chloro-2-methoxy-phenyl)-2-[4-(difluoromethyl)-2-nitrophenyl]acetate.
  • Step 3 Preparation of ethyl 2-(5-chloro-2-methoxy-phenyl)-2-[4-(difluoromethyl)-2-nitrophenyl] propanoate.
  • Step 4 Preparation of 3-(5-chloro-2-methoxy-phenyl)-6-(difluoromethyl)-3-methyl-indolin-2-one.
  • Step 1 Preparation of 3-(chloromethyl)-2-methoxy-pyridine.
  • Step 2 Preparation of 2-(2-methoxy-3-pyridyl) acetonitrile.
  • Step 3 Preparation of 2-(2-methoxy-3-pyridyl)acetic acid.
  • Step 4 Preparation of methyl 2-(2-methoxy-3-pyridyl)acetate.
  • Step 5 Preparation of methyl 2-(5-chloro-2-methoxy-3-pyridyl)acetate.
  • Step 6 Preparation of methyl 2-(5-chloro-2-methoxy-3-pyridyl)-2-[2-nitro-4-(trifluoromethyl)phenyl]acetate.
  • Step 7 Preparation of methyl 2-(5-chloro-2-methoxy-3-pyridyl)-2-[2-nitro-4-(trifluoromethyl)phenyl]propanoate.
  • Step 8 Preparation of 3-(5-chloro-2-methoxy-3-pyridyl)-3-methyl-6-(trifluoromethyl)indolin-2-one.
  • aqueous phase was extracted three times with ethyl acetate (5.0 mL).
  • the combined organic phase was dried with anhydrous Na 2 SO 4 , filtered and concentrated in vacuum.
  • the residue was purified by prep-HPLC (column: Waters Xbridge Prep OBD C18 150*40mm*10um; mobile phase: [water(0.04% NH 3 H 2 O+10mM NH 4 HCO 3 )-ACN]; B%: 65%-80%, 8min) to give 3-(5-chloro-2-methoxy-3-pyridyl)-3-methyl-6-(trifluoromethyl)indolin-2-one (65.0 mg, 65.0% yield) as a white solid.
  • Example 36 3-(5-chloro-2-methoxyphenyl)-3-methyl-6-(trifluoromethyl)-1H-pyrrolo[2,3-b]pyridin-2(3H)-one
  • Example 41 Assessment of BK channel stimulatory activity in a whole-cell patch clamp assay
  • HEK-293 cell line expressing functional alpha subunits of BK channels were passaged at a confluence of 50 to 80%. Cells were seeded into 35 mm sterile culture dishes containing 2 mL culture complete medium. Cells were cultivated at a density that enables single cells (without visible connections to neighbouring cells) to be measured.
  • test compounds (as indicated in Table 1 below) were dissolved in DMSO to achieve a stock concentration of 10 mM and stored in deep freezer (-70°C to -90°C). All test solutions were prepared shortly prior to the electrophysiological experiments and kept at room temperature (19°C to 30°C) when in use.
  • the final bath solution included the components outlined below:
  • the 1x bath solution was prepared by diluting 10x bath solution without glucose and 100x glucose solution with water at least every 7 days. Both stock solutions had been prepared prior to the experimental start of the present study and stored at 1°C to 9°C (10x bath solution) or -10°C to -30° (100x glucose solution). When in use, the 1x bath solution was kept at room temperature (19°C to 30°C). When not in use, the 1x bath solution was stored at 1°C to 9°C.
  • the 1x pipette solution was thawed every day out of a frozen 1x pipette solution, stored at -10°C to -30°C. When in use, the 1x pipette solutions were filled into the patch-pipettes and kept at room temperature (19°C to 30°C).
  • the 35 mm culture dishes upon which cells were seeded at a density allowing single cells to be recorded were placed on the dish holder of the microscope and continuously perfused (at approximately 1 mL/min) with the bath solution described. All solutions applied to cells including the pipette solution were maintained at room temperature (19°C to 30°C). After formation of a Gigaohm seal between the patch electrodes and an individual cell (pipette resistance range: 2.0 M ⁇ to 7.0 M ⁇ ; seal resistance range: >1 G ⁇ ) the cell membrane across the pipette tip was ruptured to assure electrical access to the cell interior (whole-cell patch configuration). In case the quality of the seal was poor, the process of seal formation was repeated with a different cell and a new pipette.
  • HEK cells stably expressing human BK also referred to as "KCa1.1” or KCNM1 isoform 1
  • KCa1.1 also referred to as "KCNM1 isoform 1
  • Syncropatch 384 PE (Nanion Technologies) electrophysiology testing of compounds, cells were harvested and prepared as follows. Cells were washed (1X) in Dulbecco's Phosphate Buffered Saline for approximately 30 seconds. 2 ml of cold TRP LE express was added and swirled around to cover the bottom of the flask and allowed to sit on the cells for about 3 minutes at 28 °C (approximately 90% of the cells were lifted by light tapping of the flask). 8 ml of cold media (F12 HAM's) was added to dilute the TRP LE. Cells were triturated until a single cell suspension was achieved and the cells were transferred to a 15 mL conical tube. Cell count was performed.
  • Test agents were dissolved in DMSO to give 10 mM stocks (DMSO final concentration 0.1%). Negative (0.1% DMSO) and positive (30 ⁇ M NS-1619) controls were included in each test run to assess pharmacological responsiveness.
  • HEK-293 cells stably expressing human BK (KCa1.1) were added to each well of a Nanion Syncropatch 384 well recording plate with a 4 hole/well configuration in EBSS and then a seal enhancer buffer to form electrically tight seals. Sealed cells were washed with external recording extracellular buffer two to three times while intracellular side of each well was perfused with intracellular buffer solution prior to commencement of test agent evaluation.
  • Compound evaluation comprised running the voltage protocol for ⁇ 1 minute in regular extracellular recording buffer, followed by a 10 minute application of test agent, control compound or vehicle.
  • Table 2 Effects of test compounds on BK channel activity (as determined by Syncropatch electrophysiology testing in stably transfected HEK-293 cells). Compound Effect (%) Concentration ( ⁇ M) Example 1 61 50 Example 2 46 50 Example 3 39 50 Example 4 60 50 Example 5 45 50 Example 6 89 50 Example 7 49 25 Example 8 74 50 Example 9 69 25 Example 10 27 3 Example 11 65 50 Example 12 42 50 Example 13 50 50 Example 14 31 50 Example 15 27 50 Example 17 20 25 Example 18 30 13 Example 19 24 50 Example 20 19 50 Example 21 47 13 Example 22 56 50 Example 23 37 50

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pulmonology (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Urology & Nephrology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
EP20175820.8A 2020-05-20 2020-05-20 Nouveaux ouvreurs de canaux potassiques maxi-k pour le traitement de troubles liés à l'x fragile Withdrawn EP3912625A1 (fr)

Priority Applications (11)

Application Number Priority Date Filing Date Title
EP20175820.8A EP3912625A1 (fr) 2020-05-20 2020-05-20 Nouveaux ouvreurs de canaux potassiques maxi-k pour le traitement de troubles liés à l'x fragile
CA3177423A CA3177423A1 (fr) 2020-05-20 2021-05-20 Nouveaux agents d'ouverture du canal potassique maxi-k pour le traitement de troubles associes au x fragile
BR112022023318A BR112022023318A2 (pt) 2020-05-20 2021-05-20 Abridores do canal de potássio maxi-k para o tratamento de distúrbios associados ao x frágil
IL297493A IL297493A (en) 2020-05-20 2021-05-20 Maxi-k potassium channel openers for the treatment of disorders associated with fragile x
EP21727147.7A EP4153170A1 (fr) 2020-05-20 2021-05-20 Nouveaux agents d'ouverture du canal potassique maxi-k pour le traitement de troubles associés au x fragile
AU2021274879A AU2021274879A1 (en) 2020-05-20 2021-05-20 Maxi-K potassium channel openers for the treatment of fragile X associated disorders
JP2022571242A JP2023528292A (ja) 2020-05-20 2021-05-20 脆弱X関連障害の処置のためのmaxi-Kカリウムチャネルオープナー
MX2022014461A MX2022014461A (es) 2020-05-20 2021-05-20 Nuevos abridores de canales de potasio maxi-k para el tratamiento de trastornos asociados con x fragil.
KR1020227044772A KR20230016194A (ko) 2020-05-20 2021-05-20 취약한 x 관련 장애 치료를 위한 새로운 맥시-k 칼륨 채널 개방제
PCT/EP2021/063463 WO2021234084A1 (fr) 2020-05-20 2021-05-20 Nouveaux agents d'ouverture du canal potassique maxi-k pour le traitement de troubles associés au x fragile
CN202180035813.2A CN115666554A (zh) 2020-05-20 2021-05-20 用于治疗脆性X相关病症的新型Maxi-K钾通道开放剂

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
EP20175820.8A EP3912625A1 (fr) 2020-05-20 2020-05-20 Nouveaux ouvreurs de canaux potassiques maxi-k pour le traitement de troubles liés à l'x fragile

Publications (1)

Publication Number Publication Date
EP3912625A1 true EP3912625A1 (fr) 2021-11-24

Family

ID=70802691

Family Applications (2)

Application Number Title Priority Date Filing Date
EP20175820.8A Withdrawn EP3912625A1 (fr) 2020-05-20 2020-05-20 Nouveaux ouvreurs de canaux potassiques maxi-k pour le traitement de troubles liés à l'x fragile
EP21727147.7A Pending EP4153170A1 (fr) 2020-05-20 2021-05-20 Nouveaux agents d'ouverture du canal potassique maxi-k pour le traitement de troubles associés au x fragile

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP21727147.7A Pending EP4153170A1 (fr) 2020-05-20 2021-05-20 Nouveaux agents d'ouverture du canal potassique maxi-k pour le traitement de troubles associés au x fragile

Country Status (10)

Country Link
EP (2) EP3912625A1 (fr)
JP (1) JP2023528292A (fr)
KR (1) KR20230016194A (fr)
CN (1) CN115666554A (fr)
AU (1) AU2021274879A1 (fr)
BR (1) BR112022023318A2 (fr)
CA (1) CA3177423A1 (fr)
IL (1) IL297493A (fr)
MX (1) MX2022014461A (fr)
WO (1) WO2021234084A1 (fr)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023147597A2 (fr) * 2022-01-31 2023-08-03 The Board Of Regents Of The University Of Texas System Modulateurs allostériques de sting et méthodes d'utilisation

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5565483A (en) 1995-06-07 1996-10-15 Bristol-Myers Squibb Company 3-substituted oxindole derivatives as potassium channel modulators
JP2000191661A (ja) * 1998-12-25 2000-07-11 Mitsubishi-Tokyo Pharmaceuticals Inc 環状アミド化合物
WO2001074775A1 (fr) 2000-04-03 2001-10-11 Sanofi-Synthelabo Derives d'indolin-2-one et leur utilisation en tant que ligands des recepteurs de l'ocytocine
WO2002000217A1 (fr) 2000-06-29 2002-01-03 Neurosearch A/S Utilisation de derives d'oxindole 3-substitue comme modulateurs du canal potassique kcnq
WO2002032419A2 (fr) 2000-10-17 2002-04-25 Wyeth Methodes permettant de moduler la fonction vesicale
WO2002066426A2 (fr) 2001-02-20 2002-08-29 Bristol-Myers Squibb Company Derives de fluoro oxindole en tant que modulateurs de canaux potassium kcnq
WO2003008407A2 (fr) * 2001-07-17 2003-01-30 Sanofi-Synthelabo Derives de 1-phenylsulfonyl-1,3-dihydro-2h-indol-2-one, leur preparation et leur application en therapeutique
WO2003080047A1 (fr) 2002-03-20 2003-10-02 Bristol-Myers Squibb Company Promedicaments phosphates de fluorooxindoles
WO2005080335A1 (fr) * 2004-02-13 2005-09-01 President And Fellows Of Harvard College Oxindoles 3-3-di-substitues utilises en tant qu'inhibiteurs de l'initiation de la traduction
WO2005097107A2 (fr) * 2004-04-08 2005-10-20 Topotarget A/S Composes de diphenyl ox-indol-2-on et leur utilisation dans le traitement du cancer
WO2008129075A1 (fr) * 2007-04-24 2008-10-30 Topotarget A/S Composés 3-(4-hydroxyphényl)-indolin-2-one substitués
WO2009056707A2 (fr) * 2007-08-16 2009-05-07 Sanofi-Aventis Dérivés de l'ind0l-2-0ne disubstitues en 3, leur preparation et leur application en thérapeutique
WO2009071687A1 (fr) * 2007-12-07 2009-06-11 Abbott Gmbh & Co. Kg Dérivés oxindoliques substitués par amidométhyle et leur utilisation dans le traitement de maladies vasopressine-dépendantes
WO2013001412A1 (fr) 2011-06-27 2013-01-03 Centre National De La Recherche Scientifique Compositions pour le traitement du syndrome du x fragile
WO2014154760A1 (fr) 2013-03-27 2014-10-02 Boehringer Ingelheim International Gmbh Analogues d'indoline en tant qu'inhibiteurs de brd4
WO2020183307A1 (fr) * 2019-03-08 2020-09-17 The University Of Buea Dihydro-spiro[indoline-3:1'-isoquinoléin]-2-ones et leurs analogues et dérivés et méthodes de traitement du cancer et d'autres maladies

Patent Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5565483A (en) 1995-06-07 1996-10-15 Bristol-Myers Squibb Company 3-substituted oxindole derivatives as potassium channel modulators
EP0747354A1 (fr) * 1995-06-07 1996-12-11 Bristol-Myers Squibb Company Dérivés de l'oxindole 3-substitués comme modulateurs des canaux de potassium
US5602169A (en) 1995-06-07 1997-02-11 Bristol-Myers Squibb Company 3-substituted oxindole derivatives as potassium channel modulators
JP2000191661A (ja) * 1998-12-25 2000-07-11 Mitsubishi-Tokyo Pharmaceuticals Inc 環状アミド化合物
US20070203184A1 (en) 2000-04-03 2007-08-30 Sanofi-Aventis Novel Indolin-2-one Derivatives, Their Preparation and the Pharmaceutical Compositions Comprising Them
WO2001074775A1 (fr) 2000-04-03 2001-10-11 Sanofi-Synthelabo Derives d'indolin-2-one et leur utilisation en tant que ligands des recepteurs de l'ocytocine
WO2002000217A1 (fr) 2000-06-29 2002-01-03 Neurosearch A/S Utilisation de derives d'oxindole 3-substitue comme modulateurs du canal potassique kcnq
WO2002032419A2 (fr) 2000-10-17 2002-04-25 Wyeth Methodes permettant de moduler la fonction vesicale
WO2002066426A2 (fr) 2001-02-20 2002-08-29 Bristol-Myers Squibb Company Derives de fluoro oxindole en tant que modulateurs de canaux potassium kcnq
WO2003008407A2 (fr) * 2001-07-17 2003-01-30 Sanofi-Synthelabo Derives de 1-phenylsulfonyl-1,3-dihydro-2h-indol-2-one, leur preparation et leur application en therapeutique
WO2003080047A1 (fr) 2002-03-20 2003-10-02 Bristol-Myers Squibb Company Promedicaments phosphates de fluorooxindoles
WO2005080335A1 (fr) * 2004-02-13 2005-09-01 President And Fellows Of Harvard College Oxindoles 3-3-di-substitues utilises en tant qu'inhibiteurs de l'initiation de la traduction
WO2005097107A2 (fr) * 2004-04-08 2005-10-20 Topotarget A/S Composes de diphenyl ox-indol-2-on et leur utilisation dans le traitement du cancer
WO2008129075A1 (fr) * 2007-04-24 2008-10-30 Topotarget A/S Composés 3-(4-hydroxyphényl)-indolin-2-one substitués
WO2009056707A2 (fr) * 2007-08-16 2009-05-07 Sanofi-Aventis Dérivés de l'ind0l-2-0ne disubstitues en 3, leur preparation et leur application en thérapeutique
WO2009071687A1 (fr) * 2007-12-07 2009-06-11 Abbott Gmbh & Co. Kg Dérivés oxindoliques substitués par amidométhyle et leur utilisation dans le traitement de maladies vasopressine-dépendantes
WO2013001412A1 (fr) 2011-06-27 2013-01-03 Centre National De La Recherche Scientifique Compositions pour le traitement du syndrome du x fragile
WO2014154760A1 (fr) 2013-03-27 2014-10-02 Boehringer Ingelheim International Gmbh Analogues d'indoline en tant qu'inhibiteurs de brd4
WO2020183307A1 (fr) * 2019-03-08 2020-09-17 The University Of Buea Dihydro-spiro[indoline-3:1'-isoquinoléin]-2-ones et leurs analogues et dérivés et méthodes de traitement du cancer et d'autres maladies

Non-Patent Citations (40)

* Cited by examiner, † Cited by third party
Title
"Remington: The Science and Practice of Pharmacy", PHARMACEUTICAL PRESS
ABDERRAHMAN EL BOUAKHER ET AL: "A General and Efficient Method to Access Tetracyclic Spirooxindole Derivatives : A General and Efficient Route to Tetracyclic Spirooxindole Derivatives", EUROPEAN JOURNAL OF ORGANIC CHEMISTRY, vol. 2015, no. 3, 1 January 2015 (2015-01-01), DE, pages 556 - 569, XP055769874, ISSN: 1434-193X, DOI: 10.1002/ejoc.201403292 *
ATZRODT J ET AL., BIOORG MED CHEM, vol. 20, no. 18, 2012, pages 5658 - 67
B. NARENDRAPRASAD REDDY ET AL: "Synthesis of Functionalized 6-Hydroxy-2-oxindole Derivatives by Phenoxide Cyclization", ORGANIC LETTERS, vol. 18, no. 24, 16 December 2016 (2016-12-16), US, pages 6264 - 6267, XP055743440, ISSN: 1523-7060, DOI: 10.1021/acs.orglett.6b03048 *
DALVIE D ET AL.: "Influence of aromatic rings on ADME properties of drugs", 2010, ROYAL SOCIETY OF CHEMISTRY, article "Metabolism, Pharmacokinetics and Toxicity of Functional Groups", pages: 275 - 327
DENG PY ET AL., J PHYSIOL, vol. 594, 2016, pages 83 - 97
DENG PY ET AL., NEURON, vol. 77, 2013, pages 696 - 711
DEY C ET AL., CHEM COMMUN, vol. 48, no. 25, 2012, pages 3064 - 6
EDWARD RICHMOND ET AL: "An asymmetric pericyclic cascade approach to 3-alkyl-3-aryloxindoles: generality, applications and mechanistic investigations", ORGANIC & BIOMOLECULAR CHEMISTRY, vol. 13, no. 6, 1 January 2015 (2015-01-01), pages 1807 - 1817, XP055743659, ISSN: 1477-0520, DOI: 10.1039/C4OB02526A *
EDWARD RICHMOND ET AL: "Asymmetric Pericyclic Cascade Approach to Spirocyclic Oxindoles", ORGANIC LETTERS, vol. 14, no. 11, 22 June 2012 (2012-06-22), US, pages 2762 - 2765, XP055769741, ISSN: 1523-7060, DOI: 10.1021/ol300982f *
EVANS DC ET AL., CHEM RES TOXICOL, vol. 17, no. 1, 2004, pages 3 - 16
GECZ J, ANN HUM GENET, vol. 64, 2000, pages 95 - 106
GRIBKOFF VK ET AL., NAT MED, vol. 7, 2001, pages 471 - 7
GRIGSBY J, CLIN NEUROPSYCHOL, vol. 30, 2016, pages 815 - 33
HAGERMAN PJ ET AL., ANN N Y ACAD SCI, vol. 1338, no. 1, 2015, pages 58 - 70
HALL DA ET AL., HANDB CLIN NEUROL, vol. 147, 2018, pages 377 - 91
HEBERT B ET AL., ORPHANET J RARE DIS, vol. 9, 2014, pages 124
HEWAWASAM P ET AL., BIOORG MED CHEM LETT, vol. 12, no. 7, 2002, pages 1023 - 6
HUNTER JE ET AL.: "GeneReviews", 1998, UNIVERSITY OF WASHINGTON, article "FMR1 Disorders"
JACQUEMONT S ET AL., EUR J HUM GENET, vol. 19, no. 9, 2011
JENSEN BS, CNS DRUG REV, vol. 8, no. 4, 2002, pages 353 - 60
K M SYTNIK ET AL: "Use of acidochromatic cyclocondensation in the synthesis of 2-oxo-3,3-diphenyl-2,3-dihydro-1H-thieno[3,4-b]pyrrole-6-carboxylic acid derivatives", ZHURNAL ORGANICHNOI TA FARMATSEVTICHNOI KHIMII, vol. 5, no. 1, 31 December 2007 (2007-12-31), pages 21 - 26, XP009525265 *
KINTHADA LK ET AL., ORG BIOMOL CHEM, vol. 12, no. 41, 2014, pages 8152 - 73
KUNDIG EP ET AL., ANGEW CHEM INT ED ENGL, vol. 46, no. 44, 2007, pages 8484 - 7
LATORRE R ET AL., BIOL RES, vol. 39, no. 3, 2006, pages 385 - 401
LAUMONNIER F ET AL., AM J PSYCHIATRY, vol. 163, 2006, pages 1622 - 9
LAWRENCE N J ET AL: "Synthesis of Diaryl Acetates and Oxoindoles via a Sequential VNSAR-SNAR Three-Component Coupling reaction", ORGANIC LETTERS, AMERICAN CHEMICAL SOCIETY, US, vol. 6, no. 26, 24 November 2004 (2004-11-24), pages 4957 - 4960, XP002380787, ISSN: 1523-7060, DOI: 10.1021/OL047890Y *
LIANG L ET AL., HUM MOL GENET, vol. 28, no. 17, 2019, pages 2937 - 51
LIU L ET AL., ORG LETT, vol. 13, no. 7, 2011, pages 1666 - 9
LOZANO R ET AL., INTRACTABLE RARE DIS RES, vol. 3, no. 4, 2014, pages 134 - 46
MADDALENA A ET AL., GENET MED, vol. 3, no. 3, 2001, pages 200 - 5
MARVIN W. BARKER ET AL: "Heterocycles from ketenimines. 12. Pyrrolo[3,2-b]pyridines", JOURNAL OF ORGANIC CHEMISTRY, vol. 44, no. 7, 1 March 1979 (1979-03-01), Washington, pages 1175 - 1177, XP055769883, ISSN: 0022-3263, DOI: 10.1021/jo01321a037 *
MODVIG A ET AL., J ORG CHEM, vol. 79, 2014, pages 5861 - 8
MONAGHAN KG ET AL., GENET MED, vol. 15, no. 7, 2013, pages 575 - 86
MULLEY JC ET AL., J MED GENET, vol. 32, no. 3, 1995, pages 162 - 9
MYRICK LK ET AL., PROC NATL ACAD SCI USA, vol. 112, no. 4, 2015, pages 949 - 56
RILEY C ET AL., PEDIATRICS, vol. 139, 2017, pages S147 - S52
THURMAN AJ ET AL., RES DEV DISABIL, vol. 35, 2014, pages 1072 - 86
WILLIAM JS ET AL., JOURNAL OF LABELLED COMPOUNDS AND RADIOPHARMACEUTICALS, vol. 53, no. 11-12, 2010, pages 635 - 44
WURTZ S ET AL., J AM CHEM SOC, vol. 131, no. 24, 2009, pages 8344 - 5

Also Published As

Publication number Publication date
CA3177423A1 (fr) 2021-11-25
AU2021274879A1 (en) 2022-12-08
EP4153170A1 (fr) 2023-03-29
BR112022023318A2 (pt) 2022-12-20
IL297493A (en) 2022-12-01
MX2022014461A (es) 2023-01-05
KR20230016194A (ko) 2023-02-01
WO2021234084A1 (fr) 2021-11-25
CN115666554A (zh) 2023-01-31
JP2023528292A (ja) 2023-07-04

Similar Documents

Publication Publication Date Title
US20180318255A1 (en) Pro-Neurogenic Compounds
TW202214589A (zh) Shp2抑制劑及其應用
CN104470898B (zh) 环烷基甲酸类衍生物、其制备方法及其在医药上的应用
KR102479356B1 (ko) NR2B의 N-알킬아릴-5-옥시아릴-옥타히드로-시클로펜타[c]피롤 음성 알로스테릭 조정제
AU2012388221A1 (en) Pro-neurogenic compounds
KR20050021494A (ko) 5-하이드록시트립타민-6 리간드로서의1-헤테로사이클릴알킬-3-설포닐인돌 또는 -인다졸 유도체
US11008323B2 (en) Substituted tricyclic 1,4-benzodiazepinone derivatives as allosteric modulators of group II metabotropic glutamate receptors
EA013748B1 (ru) Производные n-(гетероарил)-1-гетероарилалкил-1h-индол-2-карбоксамидов, их получение и их применение в терапии
CN101730680A (zh) 具有5-ht6受体亲和力的4’取代的化合物
TW200810752A (en) Modulators of muscarinic receptors
AU2021274879A1 (en) Maxi-K potassium channel openers for the treatment of fragile X associated disorders
JP2022511236A (ja) 置換キナゾリノン誘導体、及びmGluR4のポジティブアロステリック調節剤としてのその使用
KR101522803B1 (ko) Kat ii 억제제
CN112969692A (zh) 用于治疗多汗症的布美他尼衍生物
TW200924765A (en) Piperidine derivative
KR20220118483A (ko) Oga 억제제 화합물
JP6837578B2 (ja) 発明の名称アミノアルコール誘導体、その医薬組成物および使用
EP4010328A1 (fr) Composés de 5-hétéroaryl-pyridin-2-amine en tant qu'antagonistes du récepteur du neuropeptide ff
EP4340843A1 (fr) Inhibiteurs de slc16a3 à base de 1,2,4-triazolo[1,5-a]pyrimidine et leur utilisation thérapeutique
CN117285484A (zh) 抑制hdac6酶的苯并噻二嗪1,1-二氧化物类化合物及其制备方法和应用
WO2022243574A1 (fr) Inhibiteurs de slc16a3 à base de 3-(phtalazin-1-yl) benzènesulfonamide et leur utilisation thérapeutique
WO2024056678A1 (fr) Nouveaux promédicaments nootropiques de phénéthylamine
TW202400149A (zh) 經噻唑并芳基-甲基取代之環狀肼-n-甲醯胺衍生物

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN PUBLISHED

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

B565 Issuance of search results under rule 164(2) epc

Effective date: 20210210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20220525